EP2490713A2 - Krebsimmuntherapie und behandlungsverfahren - Google Patents

Krebsimmuntherapie und behandlungsverfahren

Info

Publication number
EP2490713A2
EP2490713A2 EP10773491A EP10773491A EP2490713A2 EP 2490713 A2 EP2490713 A2 EP 2490713A2 EP 10773491 A EP10773491 A EP 10773491A EP 10773491 A EP10773491 A EP 10773491A EP 2490713 A2 EP2490713 A2 EP 2490713A2
Authority
EP
European Patent Office
Prior art keywords
immunogen
patient
cancer
epitope
disease
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP10773491A
Other languages
English (en)
French (fr)
Inventor
Adrian Ion Bot
Zhiyong Qiu
David C. Diamond
Mihail Obrocea
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Mannkind Corp
Original Assignee
Mannkind Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Mannkind Corp filed Critical Mannkind Corp
Publication of EP2490713A2 publication Critical patent/EP2490713A2/de
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001184Cancer testis antigens, e.g. SSX, BAGE, GAGE or SAGE
    • A61K39/001189PRAME
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00119Melanoma antigens
    • A61K39/001191Melan-A/MART
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001193Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; PAP or PSGR
    • A61K39/001195Prostate specific membrane antigen [PSMA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule

Definitions

  • the instant disclosure relates to strategies for the design and practice of immunotherapy protocols for generating an effector T cell response against a target antigen in a subject, and for more advantageously utilizing immunogenic products that induce, promote the growth of, activate, and/or otherwise stimulate effector T cells targeting tumor antigens in the treatment of cancer. More particularly, embodiments relate to determining a course of treatment and methods for treating a subject wherein the presence or absence of the baseline immunoreactivity to at least one target antigen is assessed to select subjects for administration of one or more active immunotherapeutics targeting the at least one target antigen.
  • the strategies disclosed herein include selecting patients with primarily lymphatically confined disease or disease that has limited progression beyond, or is otherwise confined to, the lymphatic system for treatment with an active immunotherapeutic that is administered intralymphatically.
  • Immunotherapies are designed to help the immune system recognize cancer cells, and/or to strengthen a response against cancer cells in order to destroy the cancer.
  • Immunotherapies include active and passive immunotherapies. Active immunotherapies attempt to stimulate the body's own immune system to fight the disease. Passive immunotherapies generally do not rely on the patient's immune system to attack the disease; instead, they use immune system components (such as antibodies) generated outside of the patient's body.
  • the instant disclosure relates to methods for treating a cancer patient.
  • Some embodiments of the invention relate to methods of treating a cancer patient with an active immunotherapeutic, wherein the methods include choosing an immunotherapeutic regimen based on a patient's level of pre-existing immunoreactivity to at least one target antigen, wherein the regimen includes administration of at least one immunogen targeting the at least one target antigen, or an epitope thereof; and treating the patient according to the regimen.
  • the methods described herein include choosing an immunotherapeutic regimen targeting at least one target antigen based on a patient's known level of pre-existing immunoreactivity to the at least one antigen.
  • the methods include choosing an immunotherapeutic regimen targeting at least one target antigen based on the presence or absence of immunoreactivity the at least one target antigen in the patient. In some embodiments, the methods include choosing an immunotherapeutic regimen targeting at least one target antigen based on the patient having a substantial or minimal level of immunoreactivity to the at least one target antigen.
  • Some embodiments include a method of treating a cancer patient with an active immunotherapeutic including assessing a patient's level of pre-existing immunoreactivity to at least one target antigen; choosing an immunotherapeutic regimen based on the level of preexisting immunoreactivity, wherein the regimen includes administration of at least one immunogen targeting the at least one target antigen, or an epitope thereof; and treating the patient according to the regimen.
  • the immunogen can promote an effector T cell response to an antigen associated with the cancer.
  • the immunotherapeutic regimen can achieve a clinical benefit.
  • the clinical benefit can include tumor regression or stabilization of disease, and the like, or a combination thereof.
  • the level of pre-existing immunoreactivity can be measured by tetramer or ELISPOT assay, and the like, or a combination thereof.
  • the patient can have a cancer that has not progressed beyond secondary lymphatic organs.
  • the patient can have stage IIIC or IV (Mia) lymphatic disease.
  • the cancer can be at least one of, for example, melanoma, kidney, breast, pancreas, prostate, colorectal, ovarian, non-small-cell-lung, glioblastoma, ocular melanoma, hormone sensitive carcinoma of breast, prostate, and ovary, hormone refractory prostate carcinoma, renal cell carcinoma, esophageal, or mesothelioma, and the like.
  • the patient can be HLA-A2 positive.
  • the immunotherapeutic regimen for treating a cancer patient can include a prime-boost regimen. In some embodiments, the immunotherapeutic regimen can include more than one therapeutic cycle.
  • the immunogen can be administered direct delivery to the lymphatic system of the patient. The direct delivery to the lymphatic system can include intranodal delivery.
  • the prime -boost immunotherapeutic regimen can include administration of an effective amount of a plasmid to induce an immune response followed by administration of an effective amount of at least one peptide corresponding to an epitope expressed by the plasmid.
  • the plasmid can include, for example, pMEL-TYR or pPRA-PSM, and the like.
  • the epitope(s) expressed by the plasmid can include at least one of, for example, a PRAME epitope, or an analogue thereof, or a PSMA epitope, or an analogue thereof, or a Melan A epitope, or an analogue thereof, or a tyrosinase epitope, or an analogue thereof, and the like, or a combination thereof.
  • the epitope(s) expressed by the plasmid can include at least one of, for example, PRAME425-433, or an analogue thereof, or PSMA288-297, or an analogue thereof, or Melan A26-35, or an analogue thereof, or tyrosinase369-377, or an analogue thereof, and the like, or a combination thereof.
  • the epitope(s) expressed by the plasmid comprises at least one of Melan A 26 -35 A27Nva (ENvaAGIGILTV) (SEQ ID NO:2), or Melan A 26 -35 A27L (ELAGIGILTV) (SEQ ID NO:3), or PRAME425-433 L426Nva, L433Nle (SNvaLQHLIGNle) (SEQ ID NO:7), or PSMA288-297 I297V (GLPSIPVHPV) (SEQ ID NO:9).
  • ENvaAGIGILTV Melan A 26 -35 A27Nva
  • ELAGIGILTV Melan A 26 -35 A27L
  • PRAME425-433 L426Nva, L433Nle (SNvaLQHLIGNle) SEQ ID NO:7
  • PSMA288-297 I297V GLPSIPVHPV
  • the immunotherapeutic regimen can further include administering an immunopotentiator.
  • the immunogen can further include an immunopotentiator.
  • the immunopotentiator can include, for example, cytokines, chemokines, co-stimulatory molecules, transcription factors, and signal transduction factors, and the like, or a combination thereof.
  • the immunotherapeutic regimen can further include administering an agent to reduce the immunosuppressive nature of the tumor micro-environment to promote a clinical benefit.
  • the target antigen includes Melan A and the patient has a pre-existing immunoreactivity to Melan A.
  • the immunotherapeutic regimen can include administering an immunogen to promote an effector T cell response to Melan A, or one or more epitopes thereof.
  • the patient can have a skin and/or lymphatic disease.
  • the skin and/or lymphatic disease can be, for example, stage IIIC or IV (Mi a) lymphatic disease.
  • the immunotherapeutic regimen can include targeting Melan A26-35 epitope, or an analogue thereof.
  • the cancer can be, for example, melanoma or glioblastoma.
  • the target antigen includes at least one of PRAME or
  • the immunotherapeutic regimen can include administering the immunogen to promote an effector T cell response to the at least one of PRAME or PSMA.
  • PRAME and PSMA can be co-expressed in the patient's tumor tissue.
  • the immunotherapeutic regimen can include targeting at least one of a PRAME425-433 epitope or a PSMA288-297 epitope.
  • the cancer can be prostate, kidney cancer, or melanoma.
  • the cancer can be prostate cancer and the clinical benefit can include, for example, decreased PSA levels, and the like.
  • the patient may have no or minimal pre-existing immunoreactivity to both PRAME and PSMA; and the immunotherapeutic regimen can further include a subsequent therapy comprising: administering at least one further therapeutic cycle of an active immunotherapeutic targeting at least PRAME or PSMA according to the immunotherapeutic regimen; assaying for expansion of PRAME and/or PSMA T cells in a patient sample; classifying the patient as a responder based on the expansion of antigen specific T cells; and administering at least one further therapeutic cycle to said responder.
  • the assaying step shows an expansion in PRAME or PSMA T cells and the subsequent therapy can include repeating the therapeutic cycles of the immunotherapeutic regimen.
  • the subsequent therapy can further comprise administering an immunopotentiator.
  • the assaying step indicates no expansion or a temporary expansion of both PRAME and PSMA T cells and the subsequent therapy can include, for example, discontinuation of the immunotherapeutic regimen.
  • Some embodiments include a method of treating a cancer patient including: selecting a patient at a stage of disease wherein there is limited spread beyond the lymphatic system and any metastases that have spread beyond the lymphatic system number 10 or fewer and are either 1) not in a vital organ or 2) less than one centimeter in diameter; and applying to the patient an immunotherapeutic regimen including administering directly to the lymphatic system of the patient an immunogen to promote an effector T cell response to an antigen associated with the cancer.
  • the immunotherapeutic regimen can achieve a clinical benefit.
  • the clinical benefit can include tumor regression or stabilization of disease, and the like, or a combination thereof.
  • the patient can have a cancer that has not progressed beyond secondary lymphatic organs.
  • the patient can have stage IIIC or IV (Mia) lymphatic disease.
  • the cancer can be at least one of, for example, melanoma, kidney, breast, pancreas, prostate, colorectal, ovarian, non-small-cell-lung, glioblastoma, ocular melanoma, hormone sensitive carcinoma of breast, prostate, and ovary, hormone refractory prostate carcinoma, renal cell carcinoma, esophageal, or mesothelioma, and the like.
  • the patient can be HLA-A2 positive.
  • the immunotherapeutic regimen can include a prime -boost regimen.
  • the prime-boost immunotherapeutic regimen can include administration of an effective amount of a plasmid to induce an immune response followed by administration of an effective amount of at least one peptide corresponding to an epitope expressed by the plasmid.
  • the plasmid can include, for example, pMEL-TYR or pPRA-PSM, and the like.
  • the epitope(s) expressed by the plasmid can include at least one of, for example, a PRAME epitope, or an analogue thereof, or a PSMA epitope, or an analogue thereof, or a Melan A epitope, or an analogue thereof, or a tyrosinase epitope, or an analogue thereof, and the like, or a combination thereof.
  • the epitope(s) expressed by the plasmid can include at least one of, for example, PRAME425-433, or an analogue thereof, or PSMA288-297, or an analogue thereof, or Melan A26-35, or an analogue thereof, or tyrosinase369-377, or an analogue thereof, and the like, or a combination thereof.
  • the epitope(s) expressed by the plasmid comprises at least one of Melan A 26 - 35 A27Nva (ENvaAGIGILTV) (SEQ ID NO: 2), or Melan A 26 -35 A27L (ELAGIGILTV) (SEQ ID NO:3), or PRAME 425 -433 L426Nva, L433Nle (SNvaLQHLIGNle) (SEQ ID NO:7), or PSMA 288 _ 297 I297V (GLPSIPVHPV) (SEQ ID NO:9).
  • the immunotherapeutic regimen can further include administering an immunopotentiator.
  • the immunogen can further include an immunopotentiator.
  • the immunopotentiator can include, for example, Toll like Receptor (TLR) ligands, endocytic-Pattern Recognition Receptor (PRR) ligands, cytokines, chemokines, co-stimulatory molecules, transcription factors, and signal transduction factors, and the like, or a combination thereof.
  • the immunotherapeutic regimen can further include administering an agent to reduce the immunosuppressive nature of the tumor micro-environment to promote a clinical benefit.
  • the immunotherapeutic regimen can include more than one therapeutic cycle.
  • the immunogen can be administered direct delivery to the lymphatic system of the patient.
  • the direct delivery to the lymphatic system can include intranodal delivery.
  • the antigen associated with the cancer includes Melan A and the patient has a pre-existing immunoreactivity to Melan A.
  • the patient can have a skin and/or lymphatic disease.
  • the skin and/or lymphatic disease can be, for example, stage IIIC or IV (Mia) lymphatic disease.
  • the immunotherapeutic regimen can include targeting Melan A 2 6-35 epitope or an analogue thereof.
  • the cancer can be, for example, melanoma or glioblastoma.
  • the antigen associated with the cancer includes at least one of PRAME or PSMA and the patient has no or minimal pre-existing immunoreactivity to at least one of PRAME or PSMA. PRAME and PSMA can be co-expressed in the patient's tumor tissue.
  • the immunotherapeutic regimen can include targeting at least one of a PRAME 42 5_433 epitope or a PSMA 2 88- 2 9 7 epitope.
  • the cancer can be, for example, prostate, kidney cancer, or melanoma.
  • the cancer is prostate cancer and the clinical benefit can include, for example, decreased PSA levels, and the like.
  • the patient may have no or minimal pre-existing immunoreactivity to both PPvAME and PSMA; and the immunotherapeutic regimen can further include a subsequent therapy comprising: administering at least one further therapeutic cycle of an active immunotherapeutic targeting at least PRAME or PSMA according to the immunotherapeutic regimen; assaying for expansion of PRAME and/or PSMA T cells in a patient sample; classifying the patient as a responder based on the expansion of antigen specific T cells; and administering at least one further therapeutic cycle to said responder.
  • the assaying step shows an expansion in PRAME or PSMA T cells and the subsequent therapy can include repeating the therapeutic cycles of the immunotherapeutic regimen.
  • the subsequent therapy can further comprise administering an immunopotentiator.
  • the assaying step indicates no expansion or a temporary expansion of both PRAME and PSMA T cells and the subsequent therapy can include, for example, discontinuation of the immunotherapeutic regimen.
  • Some embodiments include an immunogen for use in the treatment of a cancer in a patient, wherein the patient has a pre-existing immunoreactivity to Melan A, and wherein the immunogen is capable of promoting an effector T cell response to Melan-A.
  • the patient can be HLA-A2 positive.
  • the patient can have a skin and/or lymphatic disease, and wherein the treatment can further include determining the localization of the disease to the lymphatic organs.
  • the skin and/or lymphatic disease can be, for example, stage IIIC or IV (Mia) lymphatic disease.
  • the cancer can be, for example, melanoma or glioblastoma.
  • the treatment can include targeting a Melan A26-35 epitope.
  • Some embodiments include an immunogen for use in the treatment of a cancer in a patient, wherein the patient has no or minimal pre-existing immunoreactivity to at least one antigen selected from the group of PRAME and PSMA, and wherein the immunogen is capable of promoting a T cell effector response to the antigen for which there is no or minimal preexisting immunoreactivity.
  • the patient can be HLA-A2 positive.
  • PRAME and PSMA can be co-expressed in the patient's tumor tissue.
  • the treatment can include targeting a PRAME425-433 epitope and/or a PSMA288-297 epitope.
  • the cancer can be, for example, prostate cancer and the clinical benefit of the treatment can include, for example, decreased PSA levels, and the like, or a combination thereof.
  • the at least one target antigen can include a PRAME antigen and/or a PSMA antigen and the cancer can be, for example, prostate, kidney cancer, or melanoma.
  • the patient may have no or minimal pre-existing immunoreactivity to both PPvAME and PSMA; and the treatment can further include: administering at least one further therapeutic cycle of an active immunotherapeutic targeting at least PRAME or PSMA; assaying for expansion of PRAME and/or PSMA T cells in a patient sample; classifying the patient as a responder based on the expansion of antigen specific T cells; and administering at least one further therapeutic cycle to the responder.
  • the assaying step shows an expansion in anti-PRAME or anti-PSMA T cells and the subsequent therapy includes administering subsequent therapeutic cycles of the immunotherapeutic regimen.
  • the subsequent therapy can include, for example, administering an immunopotentiator.
  • the assaying step indicates no expansion or a temporary expansion of both PRAME and PSMA T cells and the subsequent therapy can include, for example, discontinuation of the immunotherapeutic regimen.
  • Some embodiments include an immunogen for use as a medicament in the treatment of a cancer in a patient, wherein the patient has a cancer that has not progressed, or has only limited spread, beyond secondary lymphatic organs, and wherein the immunogen is capable of promoting a T cell effector response to an antigen associated with the cancer, wherein the immunogen is for direct intralymphatic administration of the immunogen.
  • the patient can be HLA-A2 positive.
  • the patient can be at a stage of disease wherein there is limited spread beyond the lymphatic system and any metastases that have spread beyond the lymphatic system number 10 or fewer and are either 1) not in a vital organ or 2) less than one centimeter in diameter.
  • the pre-existing immunoreactivity or no or minimal immunoreactivity can be measured by, for example, tetramer or ELISPOT assay, and the like, or a combination thereof.
  • the patient can have a cancer that has not progressed, or has only limited spread, beyond secondary lymphatic organs.
  • the patient can have, for example, stage IIIC or IV (Mi a) lymphatic disease.
  • the cancer can be at least one of, for example, melanoma, kidney, breast, pancreas, prostate, colorectal, ovarian, non-small-cell-lung, glioblastoma, ocular melanoma, hormone sensitive carcinoma of breast, prostate, and ovary, hormone refractory prostate carcinoma, renal cell carcinoma, esophageal, or mesothelioma, and the like.
  • the treatment can achieve a clinical benefit.
  • the clinical benefit can include, for example, tumor regression or stabilization of disease, and the like, or a combination thereof.
  • the treatment can include a prime -boost regimen.
  • the treatment can include more than one therapeutic cycle.
  • the prime -boost immunotherapeutic regimen can include administration of an effective amount of a plasmid to induce an immune response followed by administration of an effective amount of at least one peptide corresponding to an epitope expressed by the plasmid.
  • the epitope(s) expressed by the plasmid can be at least one of, for example, a PRAME epitope or an analogue thereof, or a PSMA epitope or an analogue thereof, or a Melan A epitope or an analogue thereof, or a tyrosinase epitope or an analogue thereof, and the like, or a combination thereof.
  • the PRAME epitope can be, for example, PRAME 425 -433 or an analogue thereof.
  • the PSMA epitope can be, for example, PSMA288-297 or an analogue thereof.
  • the Melan A epitope can be, for example, Melan A26-35, or an analogue thereof.
  • the tyrosinase epitope can be, for example, tyrosinase 369 -377 or an analogue thereof.
  • the plasmid can include, for example, pMEL-TYR or pPRA-PSM.
  • the at least one peptide can include, for example, Melan A 26 -35 A27Nva (ENvaAGIGILTV) (SEQ ID NO:2) or Melan A 26 -35 A27L (ELAGIGILTV) (SEQ ID NO:3), PRAME 425 -433 L426Nva, L433Nle (SNvaLQHLIGNle) (SEQ ID NO: 7), or PSMA 288 _ 297 peptide analogue PSMA 288 _ 297 I297V (GLPSIPVHPV) (SEQ ID NO:9), and the like, or a combination thereof.
  • ENvaAGIGILTV Melan A 26 -35 A27Nva
  • ELAGIGILTV Melan A 26 -35 A27L
  • PRAME 425 -433 L426Nva, L433Nle (SNvaLQHLIGNle) SEQ ID NO: 7
  • the immunogen can be for direct delivery to the lymphatic system of the patient.
  • the direct delivery to the lymphatic system can include, for example, intranodal delivery.
  • the treatment can further include administering an immunopotentiator.
  • the immunogen can further include an immunopotentiator.
  • the immunopotentiator can include, for example, cytokines, chemokines, co-stimulatory molecules, transcription factors, signal transduction elements; agents that are involved in antigen processing and presentation, TAP 1 and TAP 2 proteins, immune or standard proteasome, beta-2- microglobulin, and MHC class I or II molecules; agents that are involved in regulating the apoptotic pathway, agents that are involved in gene control or silencing, such as DNA methylating enzymes, chromatin controlling molecules, RNA regulating molecules; Toll-like receptors (TLRs), peptidoglycans, LPS or analogues therefrom, imiquimodes, unmethylated CpG oligodeoxynuclotides (CpG ODNs); dsRNAs, bacterial dsDNA (which contains CpG motifs), and synthetic dsRNA (polyLC)
  • Figure 1 depicts a plasmid and peptide schedule of administration used in exemplary embodiments of the disclosure.
  • Figure 2 depicts a plasmid design for the pMel-pTyr plasmid and the Melan A and tyrosinase peptides for administration.
  • Figure 3 shows a 59% tumor shrinkage lasting 48+ weeks.
  • Figure 4 shows a 60% tumor shrinkage lasting 36+ weeks.
  • Figure 5 shows a 30% tumor shrinkage lasting 36+ weeks.
  • Figures 6A-6B shows Melan A MART- 1 T cells at baseline with disease stage and clinical outcome.
  • Figure 6A shows subjects with tumor response (lymphatic disease) or no tumor response at the disease stage (stage IV(Mlb, c) or stage IIIC and IV(Mla)).
  • Figure 6B shows the clinical outcome (PD, PR or SD) of subjects in Figure 6A. ( ⁇ SD for 2 cycles. $ Subjects with >1 year duration of treatment.) Closed circles represent subjects with no tumor response. Open circles represent subjects with tumor response (lymphatic disease).
  • Figure 7 shows the expansion of epitope-specific T cells in peripheral blood as measured by MHC multimer staining (tetramer assay).
  • Figure 8 depicts a plasmid design for the pPRA-PSM plasmid and the PRAME and PSMA peptides for administration.
  • Figure 9 shows a tumor shrinkage lasting 36 weeks.
  • Figure 10 shows association of clinical benefit with the expansion of epitope-specific T cells as measured by MHC multimer staining.
  • Figure 11 shows association of immune response with clinical outcome.
  • PC prostate carcinomas
  • RCC renal carcinomas
  • Mel melanomas
  • ESO Esophageal
  • Figure 12 depicts a comparison of the evaluations of tumor growth by the trial investigators (open bars) and that of an independent radiologist (black bars) showing qualitative agreement on a trend of tumor reduction or stabilization in patients with disease predominantly localized to lymph nodes and tumor progression in patients with visceral metastatic disease.
  • treatment refers to the act of treating a patient medically, such as by administration or application of remedies to a patient.
  • immunotherapeutic regimen, treatment, or protocol refers to a plan for a medical treatment or a desired course of treatment.
  • the regimens, treatments, or protocols for use in the methods described herein are therapeutic regimens for use in clinical or medical settings.
  • the regimen or protocol can specify any part of or all of the schedule of administration, the route of administration, and the specific reagents to be administered.
  • active immunotherapy refers to procedures or reagents to stimulate the body's own immune system to fight a disease.
  • Inclusive herein are active immunotherapeutic agents, compositions or products.
  • the active immunotherapeutic refers to an immunogen or an immunogenic agent or product or composition that can be used as an immunotherapeutic or therapeutic, for example by providing or aiding in treatment of disease.
  • the active immunotherapeutic agent, product or composition can provide or aid in curing of a disease.
  • the active immunotherapeutic agent, product or composition provide or aid in amelioration of a disease.
  • immunological refers to a process to induce or amplify an immune system response to an antigen. Alternatively, it refers to a process to reduce/decrease progression of cancer, or to prevent/block metastases of such a disease by the immune system. In the first definition it can connote a protective and/or therapeutic immune response, particularly proinflammatory or active immunity, though it can also include a regulatory response.
  • immunoreactivity refers to a biochemical interaction between an immune receptor and an antigen, for purposes of the instant invention particularly a T cell receptor (TCR) and a MHC/antigen complex, respectively.
  • TCR T cell receptor
  • MHC/antigen complex MHC/antigen complex
  • targeting a target antigen refers to promoting an immune response that recognizes the target antigen or at least one particular epitope thereof.
  • targeting a tumor or cell refers to promoting an immune response that recognizes an antigen or at least one particular epitope thereof expressed by the targeted tumor or cell.
  • the immune response is an effector T cell response.
  • the effector T cell response includes a cytotoxic T cell (CTL) response.
  • CTL cytotoxic T cell
  • the therapeutic effect of the active immunotherapeutic disclosed herein is mediated by effector T cells.
  • a target cell refers to a cell associated with a cancerous condition that can be acted upon by the components of the immune system, such as, for example, a neoplastic cell or a cell associated with tumor neovasculature.
  • a target cell is a cell to be targeted by the vaccines and methods disclosed herein.
  • a target cell according to this definition includes, but is not limited to, a neoplastic cell.
  • a target cell is a cell that expresses the target antigen or presents the target epitope.
  • a "target-associated antigen (TAA)” refers to a protein or polypeptide present in a target cell.
  • TuAA tumor-associated antigen
  • a TuAA refers to a TAA, wherein the target cell is a tumor-associated cell.
  • a TuAA is an antigen associated with, a neoplastic cell, or non-cancerous cells of the tumor such as tumor neovasculature or other stromal cells within the tumor microenvironment.
  • epitope refers to a site on an antigen recognized by an antibody or an antigen receptor.
  • a T cell epitope is generally a short peptide derived from a protein antigen (though non-protein antigens are also known). T cell epitopes bind to MHC molecules and are recognized by a particular T cell.
  • an epitope can include, but is not limited to, peptides presented on the surface of cells, the peptides being non-covalently bound to the binding cleft of class I MHC, such that they can interact with T cell receptors (TCR).
  • TCR T cell receptors
  • immunopotentiators or “immunopotentiating adjuvant” refers to any molecule that enhances, increases, promotes, or up-modulates the activity of the immune system or the cells thereof, and particularly their effector functions, through an interaction other than with an antigen receptor.
  • An “immunopotentiating adjuvant” refers to an adjuvant that activates professional antigen presenting cells (pAPCs) or T cells including, for example: TLR ligands, endocytic-Pattern Recognition Receptor (PRR) ligands, quillaja saponins, tucaresol, cytokines, and the like.
  • a therapeutic cycle refers to one or more priming/inducing/entraining doses followed by one or more boosting/amplifying doses. This sequence can be repeated as long as necessary to maintain a strong immune response in vivo.
  • a therapeutic cycle includes two or more priming/inducing/entraining doses followed by two or more boosting/amplifying doses.
  • a therapeutic cycle or cycle includes one priming/inducing/entraining dose followed by one or more boosting/amplifying doses.
  • one therapeutic cycle includes one or more priming/inducing/entraining dose followed by one boosting/amplifying doses.
  • the immunotherapeutic regimen, protocol or treatment disclosed herein can generally involve one or multiple cycles (such as 2 or more, 4 or more, 6 or more, 8 or more, 10 or more or so on).
  • target lesion refers to a tumor in a patient of sufficient size that it can be evaluated for change in size, especially shrinkage, in response to a cancer therapy.
  • Tumor size can be determined, for example, by tumor imaging or direct measurement. Tumor imaging can be accomplished by CT scan, MRI, and/or PET scan, and the like. Skin lesions can be directly measured. In some embodiments, target lesions measure greater than 1 cm in their largest dimension at start of treatment or evaluation.
  • non-target lesion used in contradistinction to “target lesions,” are tumors that measure less than 1 cm in their largest dimension at start of treatment or evaluation.
  • lymphatic system As used herein “lymphatically confined,” “not progressed beyond the lymphatic system” and similar constructions are to be understood in the context of the typical progression of malignant disease in which there is first local spread proximate to the site of origin either within the same tissue or organ or into adjacent tissues or organs followed by spread through the secondary lymphatic organs, particularly to lymph nodes locally or systemically and later to distant visceral sites. In some instances there are also skin or subcutaneous metastases prior to the appearance of visceral metastases.
  • lymphatically confined and/or “not progressed beyond the lymphatic system” as used herein can be used to refer to the actual limit of disease progression in a patient or the state of the disease following surgical removal and/or other interventions. Thus, these terms can refer to any point in disease progression prior to spread to the viscera. In some embodiments, these terms can be used to refer to patients at more specifically defined stages of disease as described herein. In some embodiments, a patient is characterized as having lymphatically confined disease due to the removal or ablation of visceral lesions following surgery or other treatment.
  • a patient is characterized as having disease that is "primarily lymphatically confined” or as having only “limited progression beyond the lymphatic system” (and the like) based on the presence of visceral metastases that are limited in size and/or number as compared to a pre-determined size and/or number.
  • the instant disclosure relates to a significant advancement in the utilization of active immunotherapies against carcinomas and the discovery and use of mechanistic-based, companion biomarkers, such as pre-existing immunoreactivity, to more advantageously utilize immunogenic products that induce, promote the growth of, activate, and/or otherwise stimulate effector T cells targeting tumor antigens in the treatment of cancer.
  • Exemplary advancements provided by the methods and immunogens disclosed herein include, but are not limited to, identification of patient subset(s) in which immunotherapy is predictably effective; achievement of durable objective responses with a cancer vaccine at a readily observable rate; first observation of selective potency of lymphatically administered immunotherapeutics against lymphatic metastatic disease; prognostic value of pre-existing immune status to target antigen(s) in predicting effectiveness of immunotherapy (at least against lymphatic metastatic disease); prolonged progression free survival in metastatic disease within patient subsets.
  • the disclosure relates to methods of identifying or selecting patients for an immunotherapeutic regimen that involves the administration of an agent that generates an effector T cell response against a target antigen.
  • the effector T cell response can be a CTL response.
  • the disclosure relates to a method of treating a patient having a carcinoma wherein pre-existing immunoreactivity to at least one or more target antigens is assessed in a patient prior to selection for an immunotherapeutic regimen.
  • a patient is selected for treatment with a particular immunotherapeutic or immunotherapeutic regimen on the basis of having or not having a preexisting immunoreactivity to a particular TuAA or epitope thereof.
  • selecting or identifying a patient can involve assessing the level (e.g., the presence or absence) of a pre-existing immunoreactivity to at least one target antigen— i.e. the presence or absence of T cells that recognize a target antigen, or at least one particular epitope thereof— prior to initiation of the therapeutic regimen.
  • the presence or absence of a pre-existing immunoreactivity against a target antigen can be measured, for example, by MHC multimer staining and/or ELISPOT assay, and/or the like.
  • selecting or identifying includes selecting or identifying a patient having no, or minimal pre-existing immunoreactivity to the target antigen.
  • selecting or identifying includes selecting or identifying a patient having a pre-existing immunoreactivity to the target antigen.
  • the immunoreactivity or the immunity of the patient against a target antigen is measured, and compared to a predetermined value. If the immunoreactivity is above or equal to the predetermined value, the patient is categorized as having a pre-existing immunoreactivity against the target antigen. If the immunoreactivity is below the predetermined value, the patient is categorized as having no or minimal pre-existing immunoreactivity to the target antigen.
  • the predetermined value can be chosen based on considerations including, for example, the technique employed to measure the value (for example, MHC multimer staining, and/or ELISPOT assay, and/or the like), the "normal value of immunoreactivity" against the target antigen, and the like, or a combination thereof.
  • the "normal value of the immunoreactivity" can be, for example, that of the same patient before the patient acquires the disease at issue, or the average value of healthy people (for example, people without the disease at issue) of a specific region, age group, gender, and/or the like.
  • a patient with pre-existing immunoreactivity against a target antigen has a number of antigen specific T cells at least 3 times greater than the lower limit of detection of the assay.
  • a patient lacking pre-existing immunoreactivity against a target antigen has a number of antigen specific T cells less than 3 times the lower limit of detection of the assay.
  • the lower limit of detection of the MHC multimer staining is 0.03 percent of tetramer positive CD8+ T cells per total number of CD8+ T cells.
  • Embodiments disclosed herein relate to methods of treatment wherein a patient whose disease has not spread beyond the lymphatic system is selected for treatment with a regimen that includes direct intralymphatic administration of an immunotherapeutic reagent.
  • administration is to a non-diseased lymph node.
  • the methods disclosed herein include selecting or identifying a patient having a metastatic disease that is lymphatically confined (either because the disease has not progressed beyond the lymphatic system or is confined following surgery or other treatment) or that is beyond the lymphatic system only to a minimal degree (e.g., having metastatic lesions beyond the lymphatic system but not in a vital organ, for example, but not limited to, soft tissue lesions, wherein such lesions are ten or fewer in number; or metastatic lesions not necessarily excluding those in vital organs that are less than 1 cm in diameter).
  • the methods include selecting or identifying a patient having a metastatic disease that has not progressed to the presence of visceral metastases.
  • selecting or identifying according to the methods disclosed herein includes selecting or identifying a melanoma patient having only skin lesions.
  • the methods disclosed herein include selecting or identifying a patient having metastatic disease/lesions outside of or beyond the lymphatic system.
  • such metastatic lesions that have spread beyond the lymphatic system have spread to for example, soft tissue but not to a vital organ, and the lesions are ten or fewer than ten in number. In some embodiments, there are 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 lesions.
  • the metastatic lesions beyond the lymphatic system are less than one centimeter in diameter. In some embodiments the lesions less that one centimeter in diameter can be in a vital organ.
  • the methods disclosed herein include selecting or identifying a patient having no metastases beyond the organ or tissue of disease origin, or in which there has been spread only to the adjacent organs or tissues.
  • a patient whose disease has spread beyond the lymphatic system to a non-vital organ and whose metastatic lesions are ten or fewer lesions is selected for treatment with an immunotherapeutic pursuant to the methods disclosed herein.
  • patients in general, can be selected based on the status of
  • patient selection can include being positive for a particular HLA type where the immunotherapeutic regimen generates a response restricted by that HLA type, for example, but not limited to, HLA-A2, or HLA-A*0201.
  • patients are selected according to whether a pre-existing immunoreactivity to a target antigen is substantial, present, minimal, or absent as compared to baseline or a predetermined value as discussed herein.
  • the presence or absence of a pre-existing immunoreactivity against a target antigen can be measured, for example, by MHC multimer staining, and/or ELISPOT assay, and/or the like.
  • the methods disclosed herein involve selecting a patient with a pre-existing immunoreactivity to a target antigen.
  • patients whose immunoreactivity against a target antigen is at, or above, or substantially above a predetermined value are selected for administration of an active immunotherapeutic composition targeting the target antigen or one or more epitopes thereof.
  • patients having a baseline pre-existing immunoreactivity to a target antigen that is greater than or substantially above a predetermined value for example, greater than about 0.1% of CD8 + T cells as determined by MHC multimer staining analysis are selected.
  • patients having a baseline immunoreactivity to the target antigen that is greater than a predetermined value for example, greater than about 0.10%, about 0.1 1 %, about 0.12%, about 0.13%, about 0.14%, about 0.15%, about 0.16%, about 0.17%, about 0.18%, about 0.19%, and about 0.20% (% T cells stained out of all CD8+ T cells) as determined by MHC multimer analysis are selected for administration of an active immunotherapeutic composition targeting the target antigen, or one or more epitopes thereof.
  • the methods involve selecting a patient with no, or minimal, pre-existing immunoreactivity to the target antigen.
  • patients whose immunoreactivity against a target antigen falls at or below a predetermined value are selected for administration of an active immunotherapeutic composition targeting the target antigen or one or more epitopes thereof.
  • patients having a baseline preexisting immunoreactivity to a target antigen of less than a predetermined value for example, less than about 0.1 % of CD8 + T cells as determined by MHC multimer analysis are characterized as having no, or minimal, preexisting immunity and are selected for administration of an active immunotherapeutic composition targeting the target antigen, or one or more epitopes thereof.
  • patients having a baseline immunoreactivity to the target antigen of less than a predetermined value for example, less than about 0.10%, about 0.09%>, about 0.08%>, about 0.07%, about 0.06%, about 0.05%, about 0.04%, about 0.03%, about 0.02%, or about 0.01 %, or not distinguishable from background levels as determined by MHC multimer analysis are selected for treatment.
  • a predetermined value for example, less than about 0.10%, about 0.09%>, about 0.08%>, about 0.07%, about 0.06%, about 0.05%, about 0.04%, about 0.03%, about 0.02%, or about 0.01 %, or not distinguishable from background levels as determined by MHC multimer analysis are selected for treatment.
  • the predetermined values are dependent upon the technique used to measure immunoreactivity and the response being sought. Based on the disclosure herein, these values will be apparent to those of skill in the art for a particular type of assay or measurement, as will additional or alternate classifications/stratifications useful for the methods described herein.
  • the selecting or identifying further includes selecting or identifying a patient having a metastatic disease that has not progressed beyond a predetermined stage.
  • the stage of a cancer is a descriptor of how much the cancer has spread.
  • cancers that are localized to the site or organ or tissue of origin, have invaded surrounding normal tissue, have invaded the lymphatic system, have invaded the veins and capillaries, have or have not metastasized to distant/visceral organs, for any type of carcinoma.
  • the stage often takes into account the size and/or thickness of a tumor, how deep it has penetrated the dermal layers or subcutaneous tissue, whether it has invaded adjacent organs, how many lymph nodes it has metastasized to (if any), and whether it has spread to distant organs.
  • TNM Tumor-Node-Metastasis
  • TNM Tumor-Node-Metastasis
  • N the number of lymph nodes involved
  • M metastasis to distant organs.
  • Each of these letters can include a numerical value (e.g., 0, 1 etc.) to indicate the thickness, number of lymph nodes or extent of metastasis respectively.
  • the numerical value used is, '0' if no metastasis has occurred or T if metastases are present.
  • the stages of melanoma are stage 0, 1, II, III or IV at least partially dependent on localization of the diseased tissue (for example, the tumor tissue) and in some stages can include the lymphatic organs.
  • the lymphatic organs include, for example, the spleen, lymph nodes, lymph vessels, and the like.
  • Stage 0, IA, IB, melanoma take into account the tumor size or thickness; do not involve metastasis, and are highly curable.
  • Stage IIA, IIB and IIC melanoma shows increase tumor size or thickness than observed at stage I, do not involve the lymph nodes, and is curable.
  • Stages IIIA, IIIB and IIIC melanoma refer to an advanced stage which takes into account the tumor size or thickness, whether one or more lymph nodes are involved and that the cancer has begun to metastasize.
  • Stage IV melanoma is another advanced stage which, in addition to the characteristics taken into account for Stage III melanoma, is associated with metastasis, represented by 'Ml '— distant metastasis. For example, for melanoma, stage IV M 1 a indicates that the melanoma has not progressed beyond the lymph nodes, (i.e.
  • lymphatic disease is limited to distant skin and subcutaneous tissues or lymph nodes whereas disease which has progressed beyond the lymph node to the Mlb or Ml c stage is representative of visceral disease i.e. metastasis has occurred to the lung, or any other visceral sites, or distant metastasis with elevated levels of serum lactic dehydrogenase (LDH) at any site.
  • LDH serum lactic dehydrogenase
  • the stages of renal cell carcinoma or kidney cancer are stage I, II, III or IV.
  • Stages I and II take into account the tumor size or thickness, and do not involve the metastasis.
  • Stage III renal cell carcinoma takes into account the tumor size or thickness, whether one or more nearby lymph nodes are involved, and whether the cancer has invaded the blood vessels of the kidney or the surrounding fatty tissue.
  • Stage IV renal cell carcinoma is an advanced stage which, in addition to the characteristics taken into account for Stage III of the disease, is associated with metastasis to the adrenal gland, lung, liver, bone, or brain.
  • stage I prostate cancer takes into account whether the degree of spread of the tumor is to one -half, or less, of one lobe of the prostate and that the level of PSA (prostate specific antigen) is less than 10 nanograms per milliliter.
  • PSA state specific antigen
  • Stage IIA prostate cancer the degree of spread of the tumor is similar to that of Stage I or to more than half of one lobe of the prostate, and the level of PSA is at least 10 nanograms per milliliter but less than 20 nanograms per milliliter.
  • Stage IIB prostate cancer the degree of spread of the tumor is to both lobes of the prostate, and the level of PSA is 20 nanograms per milliliter or higher.
  • Stage III prostate cancer the tumor has spread beyond the prostate to the seminal vesicles and the level of PSA varies.
  • Stage IV prostate cancer advanced stage disease, the tumor has spread beyond the seminal vesicles to nearby lymph nodes, tissues or organs such as the bladder, pelvis or rectum, or metastasized to distant lymph nodes, organs or tissues including the bone; PSA levels are varied.
  • the active immunotherapeutic product disclosed herein can be used to treat disease, such as melanoma or glioblastoma, localized to the lymph nodes.
  • the active immunotherapeutic disclosed herein is used to treat a disease stage of cancer (such as, for example, glioblastoma, melanoma, prostate, renal cell carcinoma) wherein there is limited spread of disease beyond the lymphatic system, or the disease is otherwise (predominantly) confined to the lymphatic system following surgery or other treatment, and any metastases (lesions) that have spread beyond the lymphatic system are ten or fewer in number, and are either not in a vital organ or less than one centimeter in diameter if involving a vital organ.
  • a disease stage of cancer such as, for example, glioblastoma, melanoma, prostate, renal cell carcinoma
  • metastases have spread or progressed beyond the lymphatic system but not to a vital organ (such examples of vital organs include lung, liver, adrenal gland) and such metastatic lesions are ten or fewer than ten in number.
  • the metastatic lesions have spread beyond the lymphatic system, including in some embodiments, spread to a vital organ, and such metastatic lesions are less than one centimeter in diameter.
  • the active immunotherapeutic disclosed herein can be used to treat clinically overt (macroscopic) disease, (for example, disease such as advanced melanoma), measured utilizing conventional imaging techniques such as computed tomography (CT) scanning, magnetic resonance imaging (MRI), positron emission tomography (PET), bone scan, or X-ray.
  • CT computed tomography
  • MRI magnetic resonance imaging
  • PET positron emission tomography
  • the active immunotherapeutic product can be used to treat microscopic (i.e. disease detectable by biopsy but not by imaging techniques) disease, minimal residual disease and prevent or delay disease relapse.
  • the immunotherapeutic product disclosed herein is utilized to prevent development of clinically overt metastatic lesions.
  • Exemplary immunotherapeutic regimens disclosed herein which include direct administration to the secondary lymphatic system were seen to be particularly effective against cancers that had not yet progressed beyond the lymphatic system, or had done so to only a limited extent. While intralymphatic immunization is known to be advantageous in regard to sensitivity to immunogen, for example, an advantage relating to the anatomical location of the target disease was neither predictable nor expected.
  • a patient with lymphatically confined disease or primarily lymphatically confined disease is selected for treatment under an immunotherapeutic regimen including intralymphatic administration of an immunogen targeting a tumor-associated antigen.
  • direct administration to a lymph node is utilized.
  • the cancer has not yet spread into the lymphatic system.
  • the treatment inhibits or prevents spread of the disease, for example spread of the disease beyond the lymphatic system.
  • methods to rapidly and reliably assess a patient's immune response to a component or multiple components of an active immunotherapeutic composition are used.
  • Some embodiments include defining or classifying a patient as a positive immune responder versus a non-responder.
  • the patient's immune response to at least one immunizing antigen (or target antigen) can be measured by MHC multimer staining, and/or ELISPOT assay, and/or the like, and predetermined criteria for classifying the patient as a positive immune responder can be a two-fold greater MHC multimer staining and/or three-fold greater ELISPOT reaction relative to a pretreatment baseline and significantly different from assay background.
  • Enzyme-linked immunospot is a sensitive technique for the detection of biomarkers released by cells or detection of individual cells that secrete a biomarker of interest, such as for example, cells secreting antibodies, cytokines, chemokines, or granzymes.
  • the technique is well established and correlates closely with the enzyme-linked immunosorbent assay (ELISA) technique. (Vaquerano et ah, Biotechniques. 1998 Nov; 25(5): 830-4, 836, which is hereby incorporated by reference in its entirety.)
  • a MHC multimer staining refers to the method to detect the presence of antigen-specific T cells wherein MHC-peptide complexes that have been multimerized and attached to a reporter molecule, typically a fluorescent dye, are contacted with T cells, binding to those expressing a T cell receptor (TCR) that recognizes the MHC-peptide complex and allowing their detection through the reporter molecule.
  • a reporter molecule typically a fluorescent dye
  • TCR T cell receptor
  • the multimer can include a dimer, a pentamer, a tetramer, or the like. Such reagents are commercially available.
  • an ELISPOT assay refers to the method used to assess the CD8 + CTL response by measuring IFN-gamma production by specific effector cells in an ELISPOT assay.
  • cells are immobilized on the plastic surface of a microtiter well and effector cells are added. The binding of cells by antigen-specific effector cells triggers the production of cytokines including IFN-gamma by the effector cells. The cells are then stained to detect the presence of intracellular IFN-gamma and the number of positively staining foci (spots) counted under a microscope.
  • response can be assessed based on the phenotype of antigen-specific T cells using, for example, flow cytometry to detect surface markers associated with different phenotypes. For example, an increase in the proportion of antigen-specific T cells that have an effector phenotype or an effector/memory phenotype can be taken as a positive response even in the absence of an increase in the total number of antigen specific T cells.
  • flow cytometry to detect surface markers associated with different phenotypes.
  • ICS assay intracellular cytokine staining
  • DTH response preferably an antigen-specific DTH
  • cytokine assays preferably an antigen-specific DTH
  • cell proliferation assays preferably an antigen-specific DTH
  • chromium release assays preferably an antigen-specific DTH
  • DTH response preferably an antigen-specific DTH
  • cytokine assays preferably an antigen-specific DTH
  • cell proliferation assays chromium release assays, and/or the like.
  • chromium release assays chromium release assays
  • Many technologies to carry out such assays are known in the art.
  • Such assays can be specific for a target epitope, not just the antigen, and thus can be referred to as epitope determinations.
  • Reagents that detect presentation of particular T cell epitopes from target antigens can also be used.
  • T cell lines and hybridomas include, for example, T cell lines and hybridomas, and more preferably, antibodies specific for the peptide-MHC complex and TCR multimers (see, for example, Li et ah, Nature Biotech. 23:349-354, 2005, which is incorporated herein by reference in its entirety).
  • Appropriate reagents are commercially available.
  • a patient sample such as blood, or other bodily fluids or secretions, biopsied tumor tissue, or portions thereof, such as lymphocytes or cytokines, is assayed for an immune response.
  • the immune response is measured using visual observations of the body, such as a skin test for DTH.
  • the response includes, in a non-limiting manner, tumor shrinkage or reduction or regression, tumor clearance, inhibition of tumor progression, amelioration of the cancer, minimal residual disease, clinical response (as measured by Response Evaluation Criteria in Solid Tumor (RECIST) criteria), and the like.
  • the patient is classified as a responder based on expansion of antigen specific T cells after immunization, for example after at least two cycles of an immunotherapeutic regimen and/or achievement of a clinical benefit.
  • the patient for example a prostate cancer patient, is classified as a responder based on tumor regression mirrored by PSA decline.
  • tumor responses e.g., tumor shrinkage
  • MHC multimer staining for the targeted Melan A epitope e.g., MHC multimer staining for the targeted Melan A epitope. That is, clinical outcomes and/or tumor responses were predicted by a pre-existing Melan AJ MART-1 T cell response prior to or at the time of initiation of the therapeutic regimen.
  • pre-existing immunoreactivity can be measured by other means disclosed herein and known in the art.
  • some embodiments relate to methods of treating a patient having a carcinoma, such as, melanoma, (or a cancer, such as, glioblastoma), the method including selecting a patient with a pre-existing immunoreactivity to at least one target antigen or one or more epitopes thereof, wherein the target antigen is Melan A, and administering to the patient an active immunotherapeutic composition targeting Melan A or one or more epitopes thereof.
  • the immunotherapeutic composition also targets at least one additional antigen associated with the tumor, or one or more epitopes thereof.
  • the additional tumor associated antigen is tyrosinase.
  • patients having disease confined to the lymphatic system are patients having disease confined to the lymphatic system.
  • the methods disclosed herein include selecting a patient having a carcinoma that has not progressed beyond the lymphatic system or lymph nodes.
  • a patient whose disease has spread beyond the lymphatic system but not to vital organs and whose lesions are ten or fewer in number shows clinical benefit.
  • a pre-existing immunoreactivity to Melan A has been associated with a positive clinical outcome upon treatment with an immunotherapeutic targeting Melan A.
  • a patient is selected to receive an active immunotherapeutic composition targeting Melan A based on a patient having a pre-existing immunoreactivity to Melan A.
  • a patient with preexisting immunoreactivity to a target antigen is selected for treatment with an immunotherapeutic regimen for which a positive clinical outcome is associated with such preexisting immunoreactivity.
  • the target includes Melan A and the patient has no or minimal pre-existing immunoreactivity to Melan A and the immunotherapeutic regimen includes co-administering an immunogen and an immunopotentiator to promote an immune response (e.g., an effector T cell response) to Melan A.
  • an immunogen e.g., an effector T cell response
  • the carcinoma is a Melan A positive cancer, such as, for example, melanoma or glioblastoma, and the like, and the at least one target antigen is Melan A.
  • the immunotherapeutic agent or composition can also target additional antigens associated with the tumor, for example, but not limited to, tyrosinase, or one or more epitopes thereof.
  • the immunotherapeutic regimen can involve a prime-boost protocol.
  • a particular epitope is targeted.
  • the targeted epitope includes Melan A26-35.
  • the components of the immunotherapeutic regimen are delivered separately and intranodally.
  • patients having a baseline pre-existing immunoreactivity to Melan-A target antigen of greater than a predetermined value for example, about 0.1 % of CD8 + T cells as determined by MHC multimer staining analysis are selected.
  • patients with a baseline immunoreactivity to a target antigen of greater than a predetermined value for example, greater than about 0.10%, about 0.1 1%, about 0.12%, about 0.13%, about 0.14%, about 0.15%, about 0.16%, about 0.17%, about 0.18%, about 0.19%, about 0.20% (% T cells stained out of all CD8+ T cells) as determined by MHC multimer analysis are characterized as having a pre-existing immunoreactivity to Melan-A.
  • Patients with pre-existing immunoreactivity to the target antigen of less than a predetermined value for example, less than about 0.10%, or about 0.09%, or about 0.08%, or about 0.07%, or about 0.06%, or about 0.05%, or about 0.04%, or about 0.03%, or about 0.02%, or about 0.01 % of CD8 T cells are characterized as having no or minimal immunoreactivity against Melan-A.
  • a predetermined value against which the baseline immunoreactivity is compared can be chosen based on the specific assay that is used to evaluate immunoreactivity.
  • a positive clinical outcome can be predicted by a de novo induction of a T cell response with effector cells.
  • some embodiments involve a theranostic approach (see for example U.S. Patent Application No. 1 1/155,928 (Publication No. US 2005-0287068 Al), which is hereby incorporated by reference in its entirety).
  • failure to expand or maintain a response to either antigen is used as a basis for either discontinuing treatment with the immunotherapeutic agent/composition or modifying the treatment to a more strongly immunogenic protocol, for example, to include administration of an adjuvant, including an immunopotentiating adjuvant.
  • patients having disease such as, for example, prostate cancer or renal cell carcinoma, confined to the lymphatic system show clinical benefit.
  • the methods disclosed herein include selecting a patient having a carcinoma that has not progressed beyond, or is otherwise confined to, the lymphatic system or lymph nodes.
  • a patient whose disease has spread beyond the lymphatic system but not to vital organs and involves ten or fewer than ten lesions shows clinical benefit.
  • Some embodiments relate to methods of treating a patient having a carcinoma, the methods including selecting a patient with no, or minimal, pre-existing immunoreactivity to PRAME and/or PSMA or a particular epitope of PRAME and/or PSMA, for example, as measured by MHC multimer staining, and/or ELISPOT assay, and/or the like, and administering to the patient an immunotherapeutic composition targeting PRAME and/or PSMA or one or more epitopes of one or both antigens, wherein an effector T cell response is generated against at least one target antigen or one or more epitopes of one or both antigens.
  • the immunotherapeutic composition also targets at least one additional antigen associated with the tumor or one or more epitopes thereof.
  • patients having a minimal baseline pre-existing immunoreactivity to PRAME and/or PSMA of less than a predetermined value, for example, about 0.2% of CD8 + T cells as determined by MHC multimer staining analysis are selected.
  • the carcinoma is selected from melanoma including ocular melanoma, prostate cancer including hormone sensitive and hormone refractory prostate cancer, kidney cancer such as, for example, renal cell carcinoma, pancreatic, colorectal, and the like.
  • the active immunotherapeutic can also target additional antigens associated with the tumor.
  • a clinical benefit can include, for example, but is not limited to, tumor regression
  • the target antigen includes PRAME and/or PSMA and the patient shows an immune response to at least one of the target antigens, wherein the method includes co-administering an agent to reduce the immunosuppressive tumor microenvironment to promote a clinical benefit.
  • any immunogen inducing an effector T cell response against the targeted antigens can be used, especially compositions suitable for intralymphatic delivery.
  • compositions suitable for intralymphatic delivery can be used, especially compositions suitable for intralymphatic delivery.
  • An antigen contemplated in embodiments disclosed herein is one against which a response can be mounted by the immune system of a subject, having a malignant tumor for example, to attack a tumor thereby inhibiting its growth or shrinking or reducing or eliminating it, and hence treating or curing the disease.
  • the antigen in some instances, can be matched to the specific disease found in the subject being treated, to generate an effector T cell response (also referred to as a cell-mediated immune response).
  • the immune response is a CTL (a cytotoxic T lymphocyte) response, i.e. a cytotoxic reaction by the immune system that results in lysis of the target cells (e.g., the malignant tumor cells).
  • CTL a cytotoxic T lymphocyte
  • Antigens can include, but are not limited to, proteins, peptides, polypeptides and derivatives thereof, and can also be non-peptide macromolecules.
  • the antigen is a protein.
  • the antigen is a peptide.
  • the antigen is a polypeptide.
  • the antigen is a derivative of a protein, peptide or polypeptide.
  • embodiments of the disclosure utilize peptide antigens of 8-15 amino acids in length, such as antigens of 8 amino acids in length, or antigens of 9 amino acids in length, or antigens of 10 amino acids in length, or antigens of 1 1 amino acids in length, or antigens of 12 amino acids in length, or antigens of 13 amino acids in length, or antigens of 14 amino acids in length, or antigens of 15 amino acids in length.
  • a peptide can be an epitope of a larger antigen, i.e. it is a peptide having an amino acid sequence corresponding to the site on the larger molecule that is presented by MHC/HLA molecules and can be recognized by, for example, an antigen receptor or T-cell receptor.
  • Exemplary antigens include, PRAME, PSMA (prostate-specific membrane antigen), Melan A, and tyrosinase, SSX-2, NYESO-1 , MAGE-1 , MAGE-3, BAGE, GAGE- 1 , GAGE-2, CEA, RAGE, SCP-1 , Hom/Mel-40, p53, H-Ras, HER-2/neu, BCR-ABL, E2A-PRL, H4-RET, IGH-IGK, MYL-RAR, Epstein Barr virus antigens, EBNA, human papillomavirus (HPV) antigens E6 and E7, TSP- 180, MAGE-4, MAGE-5, MAGE-6, pl 85erbB2, pl 80erbB-3, c-met, nm-23Hl , PSA, TAG-72-4, CAM 17.1 , NuMa, K-ras, ⁇ -Catenin, CDK4, Mum-1 , pl6,
  • the variant can be recognized by a response induced or stimulated by the reference molecule.
  • the reference molecule can be recognized by a response induced or stimulated by the variant.
  • Embodiments of the disclosure relate to active immunotherapeutic compositions targeting an antigen(s) expressed by a tumor, or one or more epitopes thereof, for the treatment of cancer.
  • Tumor-associated antigens can be expressed by the cancer cell itself or associated with non-cancerous components of the tumor, such as tumor-associated neovasculature or other stroma.
  • TuAAs can include antigens that are expressed on normal cells during fetal development when the immune system is immature and unable to respond, or they can be antigens that are normally present at extremely low levels on normal cells but are expressed at much higher levels on tumor cells.
  • a TuAA is an antigen associated with non-cancerous cells of the tumor, such as, for example, tumor neovasculature or other stromal cells within the tumor microenvironment. TuAA expression can be used to match a patient's cancer condition or type with an appropriate immunotherapeutic protocol or regimen.
  • bivalent or multivalent therapeutic agents are used. Such bivalent or multivalent therapeutics can target more than one antigen on a tumor cell. In instances where more than a single antigen on a tumor cell is targeted, the effective concentration of antitumor therapeutic is increased accordingly. Attack on stroma associated with the tumor, such as vasculature, can increase the accessibility of the tumor cells to the agent(s) targeting them. Thus, even an antigen that is also expressed on some normal tissue can receive greater consideration as a target antigen if the other antigens to be targeted in a bi- or multivalent attack are not also expressed by that tissue.
  • the number of tumor cells that can be recognized is maximized. Further, some tumors lose expression of a TuAA following immunization, giving rise to a resistant population. If the immune response is directed against more than one TuAA it becomes much more difficult for a resistant tumor to arise because it must then simultaneously lose expression of each of the antigens in order to escape.
  • this multivalent attack technique is employed when a tumor is positive for two, three, four or more TuAAs of the combination used.
  • immunogenic peptides corresponding to targeted epitope sequences of the antigen are contemplated herein and can include native sequence or cross- reactive analogue sequences.
  • Exemplary analogues of Melan A, tyrosinase, PRAME and PSMA epitopes are disclosed in U.S. Application Ser. No. 1 1/156,253 (Publication No. 20060063913), Ser. No. 1 1/155,929, (Publication No. 20060094661), Ser. No. 1 1/156,369 (Publication No. 20060057673), Ser. No. 1 1/454,300 (Publication No. US 2007-0060518 Al), Ser. No. 12/406,022, and U.S.
  • Cross-reactive analogue can refer to a peptide including 1-3 amino acid substitutions, and/or one amino acid deletion or addition as compared to the native peptide sequence that induces effector function (e.g., cytolysis or cytokine secretion) distinguishable from background, from a CTL reactive with the native peptide.
  • effector function is at least about 30%, about 50%, about 60%, about 70%, or about 80% of that induced by the native peptide.
  • effector function e.g., cytolysis or cytokine secretion
  • effector function is at least 30%, 50%, 60%), 70%), or 80% of that induced by the analogue peptide.
  • examples of targeted antigens and/or an analogue thereof contemplated for use in the methods disclosed herein include PRAME425-433, PSMA288-297, Melan A26-35, and Tyrosinase369-377 disclosed in Table 1 and elsewhere herein.
  • Melan A also known as MART-1 (Melanoma Antigen Recognized by T cells), is a melanin biosynthetic protein also expressed at high levels in melanomas.
  • Melan A MART- 1 is disclosed as a TuAA in U.S. Pat. Nos. 5,994,523; 5,874,560; and 5,620,886, each of which is hereby incorporated by reference in its entirety.
  • the targeted antigen is Melan A 26 -35 and/or an analogue thereof disclosed in Table 1 and elsewhere herein. Examples of Melan A analogues are disclosed in U.S. Patent No 7,605,227 which is hereby incorporated by reference in its entirety.
  • Melan-A analogs can include but are not limited to analogs of the sequence: E ⁇ A, L, Nva, or Nle ⁇ AGIGILT ⁇ V, Nva, or Nle ⁇ (SEQ ID NO: 10); or Y ⁇ M, V, Nva, or Nle ⁇ DGTMSQ ⁇ V, Nva, or Nle ⁇ (SEQ ID NO: 11); and wherein the sequence is not E ⁇ A or L ⁇ AGIGILT V (SEQ ID NO: 12) or YMDGTMSQV (SEQ ID NO:4).
  • the isolated peptide analogue of the invention may be selected from the group consisting of ELAGIGILTNva (SEQ ID NO: 13), ENvaAGIGILTV (SEQ ID NO:2), YVDGTMSQNva (SEQ ID NO: 14), YVDGTMSQV (SEQ ID NO: 15) and YMDGTMSQNva (SEQ ID NO:5).
  • the analog is an analog consisting essentially of the amino acid sequence ENvaAGIGILTV (SEQ ID NO:2).
  • the analog is an analog consisting essentially of the amino acid sequence YMDGTMSQNva (SEQ ID NO:5).
  • Tyrosinase a melanin biosynthetic enzyme
  • Tyrosinase is predominantly expressed in melanocytes with high levels often observed in melanomas.
  • Tyrosinase is considered one of the most specific markers of melanocytic differentiation. It is also expressed in glial cells, which like melanocytes, develop from the neuroectoderm. Tyrosinase is thus also a useful TuAA for glioblastomas, including glioblastoma multiform. Further details of tyrosinase as a TuAA is disclosed in U.S. Pat. No. 5,747,271 , which is hereby incorporated by reference in its entirety.
  • the targeted antigen is tyrosinase 369 -377, and/or an analogue thereof disclosed in Table 1 and elsewhere herein.
  • PRAME also known as MAPE, DAGE, and OIP4
  • CT cancer-testis
  • PRAME is known in the art as a cancer-testis (CT) antigen.
  • CT cancer-testis
  • PRAME as a TuAA is disclosed in U.S. Pat. No. 5,830,753, which is hereby incorporated by reference in its entirety.
  • the targeted antigen is PRAME 425 -433, and/or an analogue thereof disclosed in Table 1 and elsewhere herein. Examples of PRAME analogues are disclosed in U.S. Patent No 7,51 1 ,1 19 which is hereby incorporated by reference in its entirety.
  • PRAME analogs can include but are not limited to analogs including or consisting essentially of a sequence in which: P0 is X, XX, or XXX, wherein X specifies any amino acid or no amino acid; and PI is S, K, F, Y, T, Orn, or Hse; and P2 is L, V, M, I, Nva, Nle, or Abu; and P3 is L, Nva, Nle or Abu; and P4 is Q; and P5 is H; and P6 is L, Nva, Nle, or Abu; and P7 is I; and P8 is G, A, S, or Sar; and ⁇ at P9 is L, V, I, A, Nle, Nva, Abu, or L-NH 2 ; and ⁇ +l is X, XX, or XXX, wherein X specifies any amino acid or no amino acid; and wherein the sequence is not SLLQHLIGL (SEQ ID NO:6).
  • a PRAME analog can include or consist essentially of the sequence: ⁇ K, F, Y, T, Orn, or Hse ⁇ LLQHLIGL (SEQ ID NO: 16); or S ⁇ V, M, I, Nva, Nle, or AbujLQHLIGL (SEQ ID NO: 17); or SL ⁇ Nva, Nle or AbujQHLIGL (SEQ ID NO: 18); or SLLQH ⁇ Nva, Nle or AbujlGL (SEQ ID NO: 19); or SLLQHLI ⁇ A, S, or Sar ⁇ L (SEQ ID NO:20); or SLLQHLIG ⁇ V, I, A, Nle, Nva, Abu, or L-NH 2 ⁇ (SEQ ID NO:21); or ⁇ F, Y, T, Orn, or Hse ⁇ ⁇ Nva, Nle, M, or IjLQHLIGL (SEQ ID NO:22); or S ⁇ Nva, Nle, or M ⁇ LQHLIG ⁇ Nva, Nle, or
  • a PRAME analog can include or consist essentially of the sequence: ⁇ F, Y, T, Orn, or HsejLLQHLIGL (SEQ ID NO:27); or S ⁇ Nva, Nle, or MjLQHLIGL (SEQ ID NO:28); or SLLQHLIG ⁇ Nle, Nva, or L-NH 2 ⁇ (SEQ ID NO:29); or SLLQH ⁇ Nva or AbujlGL (SEQ ID NO:30); or S ⁇ Nva ⁇ LQHLIG ⁇ Nle ⁇ (SEQ ID NO:31); or ⁇ F or T ⁇ ⁇ L or Nva ⁇ LQHLIG ⁇ Nle ⁇ (SEQ ID NO:32).
  • the analog can include or consist essentially of the sequence: S ⁇ L or Nva ⁇ LQHLIG ⁇ Nle ⁇ (SEQ ID NO:33); or T ⁇ Nva ⁇ LQHLIG ⁇ Nle ⁇ (SEQ ID NO:34).
  • the analog can include or consist essentially of the sequence S ⁇ Nva ⁇ LQHLIG ⁇ Nle ⁇ (SEQ ID NO:31).
  • PSMA Prostate-specific membrane antigen
  • the targeted antigen is PSMA288-297 and/or an analogue thereof disclosed in Table 1 and elsewhere herein.
  • PSMA analogues are disclosed in U.S. Patent No 7,51 1 ,1 18 which is hereby incorporated by reference in its entirety.
  • PSMA analogs can include but are not limited to analogs including or consisting essentially of a sequence in which: P0 is X, XX, or XXX, wherein X specifies any amino acid or no amino acid; and PI is G, A, S, Abu, or Sar; and P2 is L, M, I, Q, V, Nva, Nle, or Abu; and P3 is P or W; and P4 is S; and P5 is I; and P6 is P; and P7 is V; and P8 is H; and P9 is P, A, L, S, or T; and ⁇ at P10 is I, L, V, Nva, or Nle; and ⁇ +l is X, XX, or XXX, wherein X specifies any amino acid or no amino acid; and wherein the sequence is not GLPSIPVHPI (SEQ ID NO:8).
  • the analog can include or consist essentially of the sequence: ⁇ S, Sar, or Abu ⁇ LPSIPVHPI (SEQ ID NO:35); or G ⁇ M or Nle ⁇ PSIPVHPI (SEQ ID NO:36); or G ⁇ L, I, Nva, or Nle ⁇ WSIPVHPI (SEQ ID NO:37); or GLWSIPVHP ⁇ Nva or V ⁇ (SEQ ID NO:38); or GLPSIPVH ⁇ A or S ⁇ I (SEQ ID NO:39); or GLPSIPVHP ⁇ V, L, Nva, or Nle ⁇ (SEQ ID NO:40); or G ⁇ Nle ⁇ PSIPVHP ⁇ Nva, or Nle ⁇ (SEQ ID NO:41); or G ⁇ Nva ⁇ PSIPVHP ⁇ Nva ⁇ (SEQ ID NO:42); or G ⁇ V, Nva, or Nle ⁇ PSIPVHPV (SEQ ID NO:43); or ⁇ Sar or Abu ⁇ LPSIPVHP ⁇ V or Nva ⁇ (SEQ ID
  • the analog can include or consist essentially of the sequence: ⁇ Abu ⁇ LPSIPVHPI (SEQ ID NO:51); or G ⁇ V, Nva, or AbujPSIPVHPI (SEQ ID NO:52); or GLPSIPVHP ⁇ V or Nva ⁇ (SEQ ID NO:53); or GLWSIPVHP ⁇ I or Nva ⁇ (SEQ ID NO:54); or G ⁇ Nle ⁇ PSIPVHP ⁇ Nva ⁇ (SEQ ID NO:55); or G ⁇ Nle or NvajPSIPVHPV (SEQ ID NO:56); or ⁇ A or Abu ⁇ LPSIPVHP ⁇ V or Nva ⁇ (SEQ ID NO:57); or G ⁇ Nva ⁇ WPSIPVHP ⁇ I or V ⁇ (SEQ ID NO:58); or A ⁇ Nva or NlejWSIPVHPI (SEQ ID NO:59); or A ⁇ V or NvajPSIPVHPV (SEQ ID NO:60).
  • the analog can include or consist essentially of the sequence: ⁇ Abu ⁇ LPSIPVHPI (SEQ ID NO:51); or GLPSIPVHP ⁇ V or Nva ⁇ (SEQ ID NO:53); or GLWSIPVHPI (SEQ ID NO:61); or G ⁇ Nle ⁇ PSIPVHP ⁇ Nva ⁇ (SEQ ID NO:55).
  • the analog can include or consist essentially of the sequence: GLPSIPVHPV (SEQ ID NO:9).
  • the immunotherapeutic regimen includes administration of two particular peptide analogues (E-PRA (SEQ ID NO:7) and E-PSM (SEQ ID NO:9)).
  • E-PRA refers to an analogue of PRAME 425 -433, PRAME 425 - 4 33 L426Nva L433Nle (SEQ ID NO:7)
  • E-PSM refers to an analogue of PSMA 28 8-297, PSMA 288 _ 297 I297V (SEQ ID NO:9).
  • E-PRA refers to an analogue of PRAME 425 -433, PRAME 425 - 4 33 L426Nva L433Nle (SEQ ID NO:7)
  • E-PSM refers to an analogue of PSMA 28 8-297, PSMA 288 _ 297 I297V (SEQ ID NO:9).
  • the immunotherapeutic regimen includes administration of two particular peptide analogues (E-MEL and E-TYR).
  • E-MEL refers to an analogue of Melan A 26 _35, Melan A 26 _35 A27Nva (SEQ ID NO:2)
  • E-TYR refers to an analogue of tyrosinase369-3 77 , tyrosinase369-3 77 V377Nva (SEQ ID NO:5).
  • compositions including a nucleic acid encoding an antigen or immunogenic fragment thereof.
  • some methods utilize immunotherapeutic compositions for the treatment of cancer including plasmid(s) used in combination with synthetic peptide(s).
  • the plasmid can be placed under the control of a promo ter/enhancer sequence which allows for efficient transcription of messenger RNA for the polypeptide upon uptake by antigen presenting cells (APCs).
  • APCs antigen presenting cells
  • Promoters that can be employed in embodiments disclosed herein are well known to one of ordinary skill in the art. Such promoters include, for example, viral and cellular promoters.
  • Viral promoters can include, for example, but not limited to, the cytomegalovirus (CMV) promoter, the EF1 promoter, the major late promoter from adenovirus 2 and the SV40 promoter.
  • cytomegalovirus (CMV) promoter examples include, for example, but are not limited to, the mouse metallothionein 1 promoter, elongation factor 1 alpha (EF1 -alpha), MHC Class I and II promoter, and CD3 promoter (regarding T cell specific expression, long term).
  • CMVp cytomegalovirus
  • a polyadenylation signal can be provided at the 3' end of the encoding sequence, for example a bovine growth hormone polyadenylation signal (BGH polyA), to increase messenger RNA stability and its translocation out of the nucleus and into the cytoplasm for translation.
  • BGH polyA bovine growth hormone polyadenylation signal
  • NIS nuclear import sequence
  • SV40 simian virus 40
  • the plasmid design can also include immunostimulatory motifs, for example, a CpG immunostimulatory motif can be placed in the NIS sequence and/or the plasmid backbone.
  • Immunostimulatory sequence refers to an oligodeoxyribonucleotide containing an unmethlylated CpG sequence.
  • the immunotherapeutic regimen includes a recombinant plasmid (pPRA-PSM).
  • pPRA-PSM refers to the plasmid RP12, (described in U.S. Patent Application Ser. No. 1 1/454,616 (Pub. No.
  • the immunotherapeutic composition includes a pMEL-
  • pMEL-TYR refers to the plasmid pSEM, (described in detail and referred to as pMA2M in U.S. Patent Application Ser. No. 10/292,413 (Publication No. 20030228634), entitled “EXPRESSION VECTORS ENCODING EPITOPES OF TARGET ASSOCIATED ANTIGENS AND METHODS FOR THEIR DESIGN," which is incorporated herein by reference) expressing the Melan A26-35 A27L analogue and tyrosinase369-377 epitopes (Table 1 and 2). It is understood that pMEL-TYR, pSEM and pMA2M are used interchangeably herein.
  • the pMEL-TYR plasmid encodes the Melan A and tyrosinase epitopes in a manner that allows for their expression and presentation by pAPCs.
  • immunotherapeutic products including assemblages of immunogenic compositions can be used.
  • Such assemblages can include 1, 2, or 3 plasmids as a set of individual compositions or a single composition can include two or more plasmids.
  • Such assemblages can also include multiple peptides corresponding to the epitopes expressed by the plasmids.
  • they can be provided as compositions including individual or multiple peptides.
  • an priming/inducing/entraining plasmid or plasmids will be sold together with the corresponding boosting/amplifying peptides.
  • the multiple plasmids will be sold together, but without corresponding peptides.
  • each of the assemblages above include the peptides corresponding to (that is capable of boosting/amplifying the response to) the epitopes expressed by those plasmids.
  • Some particular embodiments include an individual plasmid and one or both corresponding peptides. (Although the specific plasmids referred to herein are described as bivalent, they can also be amplified in a monovalent fashion).
  • the clinical benefit can be enhanced or augmented.
  • the immunotherapeutic regimen includes an agent that alters the tumor micro-environment so as to make it less immunosuppressive.
  • the tumor micro-environment refers to areas adjacent to and in between cancerous cells, encompassing extracellular matrix, vascular cells and other non-cancerous cells.
  • clinical benefit can be further improved— for example, in patients with less than a complete response, or in patients with visceral metastases, and the like— by combining the immunotherapeutic agent or composition with an agent that alters the tumor micro-environment so as to make it less immunosuppressive.
  • Such agents include, for example, those useful to deplete T reg cells or promote tumor inflammation and biological response modifiers (BRMs) capable of down- modulating or overcoming T reg activity, including immunopotentiators, as discussed elsewhere herein, but are not limited to such.
  • Exemplary T reg depleting agents include, for example, IL-2 fusion molecule (denileukin diphtitox), anti-CD25 antibody, cyclosphosphamide, gemcitabine, fludarabine, doxorubicin, and the like.
  • agents that alter the tumor micro- environment so as to make it less immunosuppressive include small molecules, such as, for example: cyclophosphamide, fludarabine, select tyrosine kinase inhibitors, biomolecules such as LIGHT, TLR ligands, CpG molecules, antibodies against PD- 1 , CTLA-4, TGFbeta, IL-10, and the like.
  • small molecules such as, for example: cyclophosphamide, fludarabine, select tyrosine kinase inhibitors, biomolecules such as LIGHT, TLR ligands, CpG molecules, antibodies against PD- 1 , CTLA-4, TGFbeta, IL-10, and the like.
  • An exemplary method including administration of agents to deplete Treg cells or promote tumor inflammation can be found in U.S.
  • the immunopotentiator can be an immunomodulatory molecule, such as, but not limited to, TLR ligands, endocytic-Pattern Recognition Receptor (PRR) ligands, and the like.
  • TLR endocytic-Pattern Recognition Receptor
  • PRR endocytic-Pattern Recognition Receptor
  • clinical benefit can be further improved— for example, in patients with less than a complete response, or in patients with visceral metastases, and the like— by combining at least one component of the immunotherapeutic regimen with an immunopotentiator or a biological response modifier.
  • the response is assessed following two therapeutic cycles of treatment.
  • Responses can be determined, for example, by: an increase in tetramer (MHC-multimer) positive cells, an increase in cytokine secretion (for example, secretion of a granzyme or gamma- interferon), or an increase in the proportions of effector or effector-memory T cells.
  • Assessment of response can be based on assaying T cells in peripheral blood or recovered in tumor biopsies.
  • the immunotherapeutic regimen includes an agent that alters the tumor micro-environment so as to make it less immunosuppressive.
  • clinical benefit can be further improved— for example, in patients with less than a complete response, or in patients with visceral metastases, and the like— by combining the immunotherapeutic agent/product with an agent that alters the tumor micro-environment so as to make it less immunosuppressive.
  • Example of such agents include, but are not limited to, small molecules, such as, cyclophosphamide, fludarabine, select tyrosine kinase inhibitors, and the like; biomolecules, Toll- like receptor (TLR) ligands (e.g., CpG motifs, antibodies against PD-1 , CTLA-4, TGFbeta, IL-10, and the like; and/or biomolecules, such as LIGHT, and the like.
  • small molecules such as, cyclophosphamide, fludarabine, select tyrosine kinase inhibitors, and the like
  • biomolecules e.g., CpG motifs, antibodies against PD-1 , CTLA-4, TGFbeta, IL-10, and the like
  • TLR Toll- like receptor
  • Immunopotentiators contemplated as useful in embodiments disclosed herein include, for example and in a non-limiting manner: agents that are involved in the control of an immune response such as cytokines, chemokines, co-stimulatory molecules, transcription factors, and signal transduction elements; agents that are involved in antigen processing and presentation; agents that are involved in regulating the apoptotic pathway, or agents that are involved in gene control or silencing, such as DNA methylating enzymes, chromatin controlling molecules, RNA regulating molecules, or the like are included.
  • agents that are involved in the control of an immune response such as cytokines, chemokines, co-stimulatory molecules, transcription factors, and signal transduction elements
  • agents that are involved in antigen processing and presentation agents that are involved in regulating the apoptotic pathway, or agents that are involved in gene control or silencing, such as DNA methylating enzymes, chromatin controlling molecules, RNA regulating molecules, or the like are included.
  • immunpotentiators can include, for example and in a non-limiting manner, molecules that trigger cytokine or chemokine production, such as peptidoglycans, LPS or analogues therefrom, unmethylated CpG oligodeoxynuclotides (CpG ODNs); dsR As such as bacterial dsDNA (which contains CpG motifs) and synthetic dsR A (polyLC) on APC and innate immune cells that bind to TLR9 and TLR3, respectively.
  • CpG ODNs unmethylated CpG oligodeoxynuclotides
  • dsR As such as bacterial dsDNA (which contains CpG motifs) and synthetic dsR A (polyLC) on APC and innate immune cells that bind to TLR9 and TLR3, respectively.
  • TLRs Toll-like receptors
  • PAMPs pathogen-associated molecular patterns
  • TLRs such as the purely synthetic anti-viral imidazoquinolines, e.g., imiquimod and resiquimod, that have been found to stimulate the cellular path of immunity by binding the TLRs 7 and 8 (Hemmi, H. et ah, Nat Immunol 3: 196-200, 2002; Dummer, R. et ah, Dermatology 207: 1 16-1 18, 2003; each of which is incorporated herein by reference in its entirety).
  • imiquimod and resiquimod that have been found to stimulate the cellular path of immunity by binding the TLRs 7 and 8
  • Immunopotentiators can further include immunopotentiating adjuvants that activate pAPC or T cells including, for example: endocytic-Pattern Recognition Receptor (PRR) ligands, quillaja saponins, tucaresol and the like.
  • PRR endocytic-Pattern Recognition Receptor
  • Immunopotentiators can interact directly with receptors that detect microbial components.
  • additional therapeutic molecules include, but are not limited to, transcription factors such as T-bet, STAT-1 STAT-4 and STAT-6. Additionally, molecules that act downstream in the signaling pathway can also be used.
  • the targeted molecules can include TLR and its downstream signaling molecules such as, for example and not limited to, MyD88, NFK-B and the like.
  • Cytokines are also contemplated for use in embodiments disclosed herein, such as, for example but not limited to, interferons (e.g, .IFN-gamma), IL-2, IL-10, IL- 12, IL-18 and TGF-beta GM-CSF, fit3 ligand (fit3L), TNF- alpha, and the like.
  • Costimulatory factors such as CD40 B7- 1 and B7-2 are also contemplated as useful herein.
  • Antibodies that bind to co-stimulatory molecules are also contemplated as useful in embodiments of the disclosure.
  • checkpoint proteins such as, for example but not limited to, FOXp3, B7-like molecules, LAG-3 ligands and such molecules are also contemplated as useful in some embodiments.
  • chromatin controlling molecules and R A regulating molecules are also contemplated. Proteins present in the antigen processing and presentation pathway such as, for example but not limited to, HLA and TAPs (Transporters associated with Antigen Processing- 1 and -2 (TAP1 and TAP2)), immune or standard proteasome, beta-2- microglobulin, and MHC class I or II molecules are also contemplated for use in some embodiments.
  • Dendritic cell activation suppressor SOCS 1 and proteins in the DNA methylation pathway such as DMNT1 are also contemplated as useful in embodiments disclosed herein. Proteins present in the apoptotic pathway are also contemplated as useful in embodiments disclosed herein. Additionally, chemokines, such as, for example, but not limited to, IL-8, MIP- 3-alpha, MIP- 1 -beta, MCP-1 , MCP-3, RANTES, and the like can also be employed in embodiments disclosed herein. In some embodiments, additional therapeutic molecules involved in promoting or maintaining T cells and/or expression of differentiation antigens (for example melanoma differentiation antigens) are contemplated herein for use with the active immunotherapeutic agent disclosed.
  • differentiation antigens for example melanoma differentiation antigens
  • Such therapeutic molecules include in some embodiments, inhibitors of BRAF (serine/threonine kinase B-Raf), for example, PLX4720 (see also WO2007/002325 disclosed herein for all it contains relating to BRAF inhibitors) which can promote T cell expansion or persistence, increase differentiation antigen expression levels and promote T cell activity against the cancer.
  • BRAF serine/threonine kinase B-Raf
  • PLX4720 see also WO2007/002325 disclosed herein for all it contains relating to BRAF inhibitors
  • MAPK MAPK/extracellular signal-regulated kinase
  • any immunotherapeutic regimen that generates an effector T cell response against the targeted antigen(s) or epitopes can be used in the disclosed methods. Those including a step for intralymphatic administration of an immunogen are particularly suitable. Prime -boost type protocols are also considered advantageous. One "prime-boost" protocol is discussed in U.S. Patent No. 6,663,871 entitled “Methods and reagents for vaccination which generate a CD8 T cell immune response," which is hereby incorporated by reference in its entirety.
  • the exemplary immunotherapeutic regimens applied in the methods disclosed herein are based on preclinical models utilizing an initial composition that establishes or entrains the immune response and a second composition that intensifies, augments or enhances the response to effective levels.
  • the first and second compositions are in forms that are the same, and in some embodiments, the first and second compositions are in forms that are different.
  • An "entraining" immunogen as contemplated in embodiments disclosed herein includes, in many embodiments, an induction that confers particular stability on the immune profile of the induced lineage of T cells.
  • prime -boost protocols utilizing a single form of immunogen—e.g., protein, polypeptide, peptide, plasmid, RNA, viral vector, and the like— can also be used in the methods disclosed herein.
  • immunogen e.g., protein, polypeptide, peptide, plasmid, RNA, viral vector, and the like
  • the immunotherapeutic regimen can involve a prime-boost protocol or induce-and-amplify protocol. In some embodiments, the immunotherapeutic regimen can involve an entrain-and-amplify protocol.
  • the active immunotherapeutic regimen includes a plasmid and one or more peptides or analogues thereof, which can be administered in treating a cancer in a subject using any such regimens/protocols (as disclosed in U.S. Patent Application No. 1 1/879,078 (Pub. No. 2008-001421 1) and U.S. Patent Application No. 10/871 ,707 (Pub. No. 2005-0079152), U.S. Patent Application No.
  • Some embodiments use an entrain-and-amplify therapeutic regimen to achieve a multivalent attack, offering the advantage of increasing the sensitivity of the tumor to attack. If more than a single antigen on a tumor cell is targeted, the effective concentration of antitumor agent is increased accordingly. Attack on stroma associated with the tumor, such as vasculature, can also increase the accessibility of the tumor cells to the agent(s) targeting them. Thus, even an antigen that is also expressed on some normal tissue can receive greater consideration as a target antigen if the other antigens to be targeted in a multivalent attack are not also expressed by that tissue.
  • exemplary immunotherapeutic regimens applied to the patient selected according to the methods disclosed herein involve priming with a plasmid such as, for example, pPRA-PSM or pMEL-TYR, and boosting with the respective peptide analogues, e.g., E-PRA and E-PSM, or E-MEL and E-TYR, respectively.
  • a plasmid such as, for example, pPRA-PSM or pMEL-TYR
  • boosting with the respective peptide analogues e.g., E-PRA and E-PSM, or E-MEL and E-TYR, respectively.
  • the methods can include, applying a regimen that includes, for example, 1-6 priming/inducing/entraining doses.
  • the method can include administering a plurality of priming/inducing/entraining doses, wherein said doses are administered over a course of one to about seven days.
  • the priming/inducing/entraining doses, boosting/amplifying doses, or priming/inducing/entraining and boosting/amplifying doses can be delivered in multiple pairs of injections, wherein a first member of a pair can be administered within about 4 days of a second member of the pair, and wherein an interval between first members of different pairs can be at least about 14 days.
  • An interval between a last priming/inducing/entraining dose and a first boosting/amplifying dose can be between about 7 and about 100 days, for example.
  • “boost/amplification,” as of a T cell response includes, in many embodiments, a process for increasing the number of cells, the number of activated cells, the level of activity, rate of proliferation, or similar parameter of T cells involved in a specific response.
  • the immunotherapeutic regimen or protocol can be adjusted based on the responsiveness to induction or amplification phases and variation in antigen expression. For example, repeated priming or inducing or entraining doses can be administered until a detectable response is obtained before administering boosting or amplifying dose(s), rather than boosting or amplifying after some set number of priming/inducing/entraining doses.
  • boosting/am lifying or maintenance doses of peptide can be discontinued if their effectiveness wanes, antigen-specific regulatory T cell numbers rise, or some other evidence of tolerization is observed, and further priming/inducing/entraining doses can be administered before resuming boosting/amplifying doses.
  • the integration of diagnostic techniques to assess and monitor immune responsiveness with methods of immunization is discussed more fully in Provisional U.S. Patent Application Ser. No. 60/580,964, which was filed on Jun. 17, 2004 and U.S. Patent Application Ser. No. 1 1/155,928 (Pub. No. 20050287068), filed Jun. 17, 2005, both entitled "IMPROVED EFFICACY OF ACTIVE IMMUNOTHERAPY BY INTEGRATING DIAGNOSTIC WITH THERAPEUTIC METHODS,” each of which is hereby incorporated by reference in its entirety.
  • a vector composition is injected into the inguinal lymph node followed by subsequent administration of a peptide antigen as a bolus.
  • one or more priming/inducing/entraining composition(s) and one or more boosting/amplifying composition(s) antigen are administered intralymphatically.
  • one or more priming/inducing/entraining composition(s) or one or more boosting/amplifying composition(s) antigen are administered intralymphatically.
  • one or more boosting/amplifying composition(s) antigen are administered intralymphatically.
  • one or more components can be delivered by infusion, generally over several hours to several days.
  • the immunotherapeutic regimen or protocol calls for injection or infusion into one or more lymph nodes, starting with a number of administrations (e.g., 1 to 10, or more, 2 to 8, 3 to 6, 4 or 5 etc.) of recombinant DNA (dose range of about 0.001 to about 10 mg/kg, about 0.005 to about 5 mg/kg, such as about 0.01 to about 1 mg/kg, such as about 0.1 to about 0.5 mg/kg) followed by one or more, e.g., 2 or 3 or 4 or 5 or 6 or 7 or 8 or 9 or 10 administrations of peptide, preferably in an immunologically inert vehicle or formulation (dose range of about 1 mg/kg to about 10 mg/kg, preferred about 0.005 to about 5 mg/kg, such as about 0.01 to about 1 mg/kg, such as about 0.1 to about 0.5 mg/kg).
  • a number of administrations e.g., 1 to 10, or more, 2 to 8, 3 to 6, 4 or 5 etc.
  • the preferred concentration of plasmid (or an effective amount of a plasmid) and peptide (or an effective amount of peptide) upon injection is generally about 0.1 mg/ml to about 10 mg/ml, such as about 0.5 to about 5 mg/ml, such as about 1 to about 2.5 mg/ml, or about 1 mg/ml, generally irrespective of the size or species of the subject.
  • particularly potent peptides can have optimum concentrations (effective amounts) toward the low end of this range, for example, between about 1 and about 100 ⁇ g/ml, such as between about 10 and about 50 ⁇ g/ml, or between about 20 and about 60 ⁇ g/ml, or between about 30 and about 70 ⁇ g/ml, or between about 40 and about 80 ⁇ g/ml, or between about 50 and about 90 ⁇ g/ml.
  • the time between the last priming/inducing/entraining dose of DNA and the first boosting/amplifying dose of peptide is not critical. In some embodiments, the time between priming/inducing/entraining and boosting/amplifying is about 7 days or more, such as about 2 weeks or about 3 weeks or about 1 month, and can exceed several months.
  • the multiplicity of injections of the priming/inducing/entraining doses and/or the boosting/amplifying doses can be reduced by substituting infusions lasting several days (preferred 2-7 days, such as 2 days or 3 days or 4 days or 5 days or 6 days or 7 days).
  • the immunotherapeutic regimen disclosed herein can be integrated with other traditional cancer therapies including, but not limiting to, surgery, radiation, chemotherapy, hormonal therapy and the like.
  • the immunotherapeutic regimen disclosed herein can be used in an adjuvant or neoadjuvant setting or treatment protocol to enhance or augment the clinical benefit.
  • the immunotherapeutic regimen can be incorporated into standard oncology therapy paradigms such as, adjunctive or consolidation therapy, involving surgery, radiation, chemotherapy, biotherapy, gene therapy, or hormonal therapy, and the like to enhance or augment the clinical benefit.
  • tumor ablative treatment refers to therapy or treatment prior to surgery or other subsequent therapy. That is, at least one therapeutic cycle of the immunotherapeutic regimen described herein is completed prior to a tumor ablative treatment such as, for example, but not limited to, surgery, radiation, or direct chemotherapy.
  • the tumor ablative treatment can be administered within days or two weeks of the completion of the therapeutic cycle as determined by the skilled practitioner.
  • the use of the immunotherapeutic regimen in a neoadjuvant therapeutic setting can increase the rate of complete and partial remission in patients, decrease the rate of relapse, and/or increase median disease free survival thereby improving the clinical benefit.
  • the immunotherapeutic regimen can be used in an adjuvant setting to increase the likelihood of a cure. That is, the cancer can be put into complete remission by a tumor ablative treatment such as, for example, but not limited to, surgical removal, irradiation, or chemotherapy with doses that are directly cytotoxic to the cancer cells, and the like.
  • the immunotherapeutic regimen is subsequently administered, resulting in a decreased rate of relapse and increased interval of disease-free survival.
  • the immunotherapeutic regimen can be administered within hours, days, or weeks of the completion of the initial treatment.
  • the immunotherapeutic regimen can be used as consolidation therapy. This resembles the adjuvant setting described above except that complete remission is not necessarily attained. In this setting, the immunotherapeutic regimen improves or increases the time to progression and progression-free survival (in the case of partial remission), and decreases the rate or time to relapse (in the case of complete remission).
  • the immunotherapeutic regimen can be used as adjunctive therapy, that is, in further combination with a tumor ablative treatment to increase that treatment's efficacy.
  • a tumor ablative treatment to increase that treatment's efficacy.
  • the two treatments are used together to increase the rate of response or disease control rate (that is of partial or complete remission).
  • the actual schedule of the two treatments can be similar to those above, but therapeutic cycles of the immunotherapeutic regimen can be alternated with rounds of the primary treatment such as chemotherapy or radiation.
  • surgery can be carried out during the time interval of a therapeutic cycle of the immunotherapeutic regimen, for example in the interval between the priming (induction/entraining) and boost (amplification) stages or in an interval between courses of the amplification composition.
  • the immunotherapy in a patient with skin or subcutaneous metastases, but not yet visceral metastases, the immunotherapy is used to stabilize the disease, or reduce or ablate tumor burden at the site of origin and/or in the lymphatic system, and the skin or subcutaneous tumors are surgically resected.
  • the immunotherapy in a patient with skin or subcutaneous metastases, but not yet visceral metastases, is used to stabilize the disease, or reduce or ablate tumor burden in the lymphatic system, prior to resection of the skin or subcutaneous tumors.
  • the immunotherapeutic regimen can be in a neoadjuvant setting (vaccine first, standard of care second) to stabilize the disease prior to tumor removal leading to long term remission.
  • exemplary cancers susceptible to such an approach include kidney carcinomas and melanomas.
  • the typical course of treatment includes surgical resection of tumor at the site of origin.
  • Such a course of treatment is also applicable to a variety of solid tumors particularly in, but not necessarily limited to, earlier stages of the disease. Examples include prostate and kidney cancer.
  • the immunotherapeutic regimen is applied in an adjuvant setting or as consolidation therapy, depending on the degree of spread of the disease that had already occurred.
  • the disclosure relates to methods of administering the one or more therapeutic agents of the immunotherapeutic regimen disclosed herein.
  • Such methods can include, without limitation: intralymphatic, intranodal, perinodal, oral, transdermal, intravenous, intradermal, intramuscular, intraperitoneal, mucosal administration, and the like. Administration can be in any manner compatible with the dosage formulation and in such amount as will be therapeutically effective.
  • An effective amount or dose of an immunogenic composition of the disclosure is that amount found to provide a desired response in the subject to be treated.
  • a particularly useful method of vaccine delivery to elicit a CTL response is disclosed in U.S. Patent Nos.
  • the compositions disclosed herein are administered directly to the lymphatic system or lymphatic organ.
  • this is to a lymph node.
  • Afferent lymph vessels can be similarly utilized. Choice of lymph node is not critical.
  • Inguinal lymph nodes can be utilized for their size and accessibility, but axillary and cervical nodes and tonsils can be similarly advantageous.
  • Administration to a single lymph node can be sufficient to induce or amplify an immune response.
  • administration to multiple nodes can increase the reliability and magnitude of the response. For embodiments promoting a multivalent response and in which multiple boosting/amplifying peptides are therefore used, each peptide can be administered to any lymph node on any occasion.
  • one peptide can be administered to the right inguinal lymph node and a second peptide to the left inguinal lymph node at the same time, for example.
  • Additional peptides can be administered to other lymph nodes even if they are not sites of induction, as it is not essential that initiating and boosting/amplifying doses be administered to the same site, due to migration of T lymphocytes.
  • additional peptides can be administered a few days later, for example, to the same lymph nodes used for the previously administered boosting/amplifying peptides as the time interval between induction and amplification generally is not a crucial parameter.
  • the time interval can be greater than about a week.
  • Segregation of administration of boosting/amplifying peptides is generally of less importance if their MHC-binding affinities are similar, but can grow in importance as the affinities become more disparate. Incompatible formulations of various peptides can also make segregated administration useful.
  • each of the components is directed to a lymph vessel, lymph node, the spleen, or other appropriate portion of the lymphatic system.
  • each component is administered as a bolus.
  • one or more components are delivered by infusion, generally over several hours to several days.
  • the immunotherapeutic compositions are directed to a lymph node such as an inguinal or axillary node by inserting a catheter or needle to the node and maintaining the catheter or needle throughout the delivery.
  • Suitable needles or catheters are available made of metal or plastic (e.g., polyurethane, polyvinyl chloride (PVC), TEFLON, polyethylene, and the like).
  • metal or plastic e.g., polyurethane, polyvinyl chloride (PVC), TEFLON, polyethylene, and the like.
  • the catheter or needle In inserting the catheter or needle into the inguinal node for example, the inguinal node is punctured under ultrasonographic control using a Vialon M Insyte W.TM. cannula and catheter of 24G3/4 (Becton Dickinson, USA) which is fixed using TegadermTM transparent dressing (TegadermTM, St. Paul, Minn., USA). An experienced radiologist generally does this procedure.
  • the location of the catheter tip inside the inguinal lymph node is confirmed by injection of a minimal volume of saline, which immediately and visibly increases the size of the lymph node.
  • the latter procedure allows confirmation that the tip is inside the node. This procedure can be performed to ensure that the tip does not slip out of the lymph node and can be repeated on various days after implantation of the catheter. In the event that the tip does slip out of location inside the lymph node, a new catheter can be implanted.
  • Various parameters can be taken into account in delivering or administering an immunogenic composition to a subject.
  • a dosage regimen and immunization schedule can be employed.
  • the amount of the components in the immunotherapeutic composition will vary from patient to patient and from antigen to antigen, depending on such factors as: the activity of the antigen in inducing a response; the flow rate of the lymph through the patient's system; the weight and age of the subject; the type of disease and/or condition being treated; the severity of the disease or condition; previous or concurrent therapeutic interventions; the capacity of the individual's immune system to synthesize antibodies; the degree of protection desired; the manner of administration and the like, all of which can be readily determined by the practitioner.
  • the immunotherapeutic compositions described herein can be delivered at a rate of from about 1 to about 500 microliters/hour or about 24 to about 12,000 microliters/day.
  • the concentration of the immunotherapeutic composition is such that about 0.1 micrograms to about 10,000 micrograms of the therapeutic composition will be delivered during a 24 hour period.
  • the flow rate is based on the knowledge that, in each minute, approximately about 100 to about 1 ,000 microliters of lymph fluid flows through an adult inguinal lymph node.
  • An objective is to maximize local concentration of vaccine formulation in the lymph system.
  • a certain amount of empirical investigation on patients is conducted to determine the most efficacious level or optimal level of infusion for a given immunotherapeutic in humans.
  • the immunogenic compositions described herein can be administered as a number of sequential doses. Such doses can be 2, 3, 4, or more doses as is needed to obtain the appropriate immune response. In some embodiments, it is contemplated that the doses of the immunogenic composition can be administered within about seconds or minutes of each other into the right or left inguinal lymph nodes.
  • the plasmid primarye
  • the combination of one or more plasmids expressing one or more immunogens can be administered.
  • the subsequent injection following the first injection into the lymph node can be within about 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10 or more minutes but not greater than about 30, 40, 50, or 60 minutes after the first injection. Similar considerations apply to the administration of two peptides individually to the right and left lymph nodes. It can be desirable to administer the doses of the immunogenic composition at an interval of days, for example, where 1 , 2, 3, 4, 5, 6, or 7, or more days elapse between subsequent administrations. In some embodiments, it can be desirable for subsequent administration(s) of the compositions to be administered via bilateral inguinal lymph node injection within about 1 , 2, 3, or more weeks or within about 1 , 2, 3, or more months following the initial dose administration.
  • all or a subset of the components of the immunotherapeutic compositions disclosed herein can be packaged or assembled together in a kit.
  • the therapeutic proteins, peptides, polypeptides, epitopes or nucleic acid encoding such can be packaged together, or as single molecules or as a set of molecules.
  • the compositions disclosed herein can be packaged and sold individually along with instructions, in printed form or on machine-readable media, describing how they can be used in conjunction with each other as disclosed herein, for use as a therapeutic.
  • the kit can contain, in a suitable container means, one or more therapeutic agents and instructions for employing the methods disclosed herein.
  • the kit can have a single container means, and/or it can have distinct container means for additional compounds such as an immunological or therapeutic effective formulation of one or more therapeutic agents for treating a disease or condition due to, for example, a proliferative disease such as cancer.
  • the liquid solution is an aqueous solution, with a sterile aqueous solution being particularly preferred.
  • the compositions can also be formulated as a deliverable and/or injectable composition.
  • the container means can itself be a syringe, pipette, and/or other such apparatus, from which the formulation can be delivered or injected into a subject, and/or even applied to and/or mixed with the other components of the kit.
  • the components of the kit can be provided as dried powder(s). When components (e.g., reagents) are provided as a dry powder, the powder can be reconstituted by the addition of a suitable solvent. It is envisioned that the solvent can also be provided in another container means.
  • one or more components of the kit can be provided as a liquid solution or a dry powder.
  • the plasmid can be sold together with the therapeutic protein, peptide, epitope or nucleic acid encoding such.
  • sets of therapeutic proteins, peptides, epitopes or nucleic acids encoding such can be sold together without the plasmid.
  • each of the components including the immunotherapeutic regimen disclosed herein can be sold separately.
  • the container means will generally include at least one vial, test tube, flask, bottle, syringe and/or other container means, into which the one or more therapeutic agents can be placed.
  • the kit can also include a second container means for containing a sterile, pharmaceutically acceptable buffer and/or other diluent.
  • the kit can also include a means for containing the materials for practicing the methods disclosed herein, and any other reagent containers in close confinement for commercial sale.
  • Such containers can include, for example, injection or blow-molded plastic containers into which the desired vials are retained.
  • the kit(s) described herein can also include, or be packaged with, an instrument for assisting with the injection/administration of the one or more therapeutic agents within the body of a subject.
  • an instrument can be, for example, but not limited to, a syringe, pump and/or any such medically approved delivery vehicle.
  • priming/entraining/inducing and boosting/amplifying compositions targeting a single epitope, or set of epitopes can be packaged together.
  • multiple priming/entraining/inducing compositions can be assembled in one kit and the corresponding boosting/amplifying compositions assembled in another kit.
  • compositions can be packaged and sold individually along with instructions, in printed form or on machine-readable media, describing how they can be used in conjunction with each other to achieve the beneficial results of the indicated immunization protocol or immunotherapeutic regimen. Further variations will be apparent to one of skill in the art.
  • the active immunotherapeutic described herein can be used in the manufacture of a medicament for use in the treatment of a disease, for example, a skin and/or lymphatic disease, a cancer, in a patient.
  • an active immunotherapeutic product (referred to as MKC1106-MT) encompassing three components: a recombinant plasmid expressing fragments of two targeted antigens, Melan A/MART- 1 and Tyrosinase (pMEL-TYR); and two peptides, epitope analogues corresponding to the respective target CTL epitopes Melan A/MART- 126-35 and Tyrosinase369-377 (E-MEL and E-TYR, respectively), was tested in an immunization regimen as shown in Figure 1.
  • the plasmid design and peptide analogues are as shown in Figure 2.
  • the plasmid was formulated as a sterile solution for administration at a concentration of 4.0 mg/mL.
  • the multicomponent active immunotherapeutic product was delivered by intralymph node injection using a plasmid prime/peptide boost approach aimed to elicit a functional immune response against both the Melan A/MART- 1 and tyrosinase antigens.
  • the plasmid dose for bilateral administration into inguinal lymph nodes was fixed at 1 ,200 micrograms per injection.
  • a total of eighteen patients were dosed— seven in the low dose peptide cohort and eleven in the high dose cohort.
  • the low dose peptide cohort received a 'low dose' of 100 micrograms each of the Melan A/MARTl and tyrosinase peptides per injection.
  • the high dose peptide cohort received a 'high dose' of 300 micrograms each of the Melan A/MARTl and tyrosinase peptides per injection.
  • the immune response rate defined as the percentage of patients who showed elevated numbers of antigen specific T cells in the blood upon immunization, as measured by MHC multimer staining, was achieved in 50% of all patients treated (Figure 7).
  • the tumor biopsies showed heavy infiltration with both CD8+ and CD4+ T cells.
  • Subset analysis identified patient populations in the trial - based on disease stage and baseline specific T cells - who demonstrated the best overall clinical responses.
  • 8 patients who had Melan A MART- 1 specific T cells at baseline four patients (all with lymphatic metastatic disease) achieved durable (>1 year) clinical responses, (PR or SD as measurable by RECIST, and tumor regression), while the other 4 patients (all with visceral disease) showed rapid disease progression and did not complete 2 cycles of treatment.
  • the remaining 10 patients without measurable specific T cells at baseline none achieved durable responses.
  • PRAME Preferential Antigen of Melanoma
  • PSMA Prostate Specific Membrane Antigen
  • MKC1 106-PP encompassed three components: a recombinant plasmid (pPRA-PSM) expressing fragments of the two targeted antigens; and two peptide analogues (E-PRA and E-PSM), corresponding to the respective target antigens ( Figure 8).
  • pPRA-PSM recombinant plasmid
  • E-PRA and E-PSM two peptide analogues
  • the plasmid was formulated as a sterile solution at a concentration of 4.0 mg/mL and each of the peptides, (E-PRA and E-PSM), were formulated as sterile solutions (at a concentration of 0.5 mg/mL and 1.0 mg/rnL respectively) from lyophilized powders.
  • E-PRA and E-PSM peptides
  • Each of the three components of the immuno therapeutic regimen was delivered by bilateral intra- inguinal lymph node injection in a prime-boost protocol, aimed to elicit a functional immune response against both PRAME and PSMA antigens.
  • the plasmid and peptide schedule of administration is shown in Figure 1.
  • a total of twenty-six patients with various tumor types (1 1 prostate carcinoma, 2 kidney carcinoma, 1 1 melanoma, and 2 with other cancers) were dosed: thirteen each in the low and high dose peptide cohort, respectively. Twenty-four patients completed at least one cycle of treatment and were deemed to be evaluable for immune response.
  • the two peptide doses used were: 'low dose' of 22.5 and 30 micrograms, and 'high dose' of 150 and 300 micrograms of peptide/injection, for E-PRA and E-PSM, respectively.
  • the plasmid dose for bilateral injection was fixed at 1 ,200 micrograms/injection.
  • Target expression analysis confirmed co-expression of PRAME and PSMA in nearly 100% of prostate cancer tumor samples and a majority of tumor tissues from kidney cancer and melanoma patients.
  • the best clinical outcomes include: a prostate carcinoma patient with tumor regression mirrored by PSA decline (30+ weeks), two prostate carcinoma patients with stable disease (36+ weeks) accompanied by PSA velocity change, a kidney cancer patient with no evidence of disease post-resection in neoadjuvant setting (72+ weeks) and a metastatic melanoma patient (Mlc stage) with stable disease at 72+ weeks.
  • the immune response rate defined as the percentage of patients who showed elevated numbers of antigen specific T cells in the blood upon immunization, was greater than 60% among evaluable patients.
  • the immunization regimen was well tolerated with five patients completing six cycles (nine months) of therapy. Seven patients achieved clinical response defined as 'Partial Response' (RECIST), or change in PSA doubling time or 'Stable Disease' (for at least six months): four with prostate, two with renal carcinoma and one with melanoma. Patients who mounted an immune response against both antigens, persisting throughout the first 2 cycles of therapy were more likely to show clinical benefit.
  • RECIST 'Partial Response'
  • the numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth used to describe and claim certain embodiments of the disclosure are to be understood as being modified in some instances by the term "about.” Accordingly, in some embodiments, the numerical parameters set forth in the written description and attached claims are approximations that may vary depending upon the desired properties sought to be obtained by a particular embodiment. In some embodiments, the numerical parameters should be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of some embodiments of the disclosure are approximations, the numerical values set forth in the specific examples are reported as precisely as practicable. The numerical values presented in some embodiments of the disclosure may contain certain errors necessarily resulting from the standard deviation found in their respective testing measurements.
  • the independent radiologist was aware of tumor type and the time sequence of images but all other patient information, including treatment, was blinded.
  • the independent radiologist was wholly responsible for selection of target and non-target lesions, their measurement, and assessing best response according to RECIST (v. 1.0) criteria based on digitized CT scans for all subjects from the trial for which imaging was available. (One responding subject had skin lesions only (Stage IIIC disease; see Figure 6A and 6B) and thus no CT scans had been taken.)
  • RECIST v. 1.0
  • Ptl-Pt4 and Pt6 showed objective response by RECIST criteria (the response for Pt6 were relatively short-lived) and Pt 7 to Ptl 1 had progressive disease despite treatment.
  • Patients (Pt)l to Pt4 in Figure 12 were those identified as having pre-existing immunity to Melan A and classified as having Stage IV: Ml a lymphatic disease (see Figure 6A and 6B).
  • Those patients classified as having Stage IV: Mlb or Ml c disease in Figure 6 A and 6B for having visceral target lesions include Pt 5 to Pt 1 1 in Figure 12.
  • Ptl to Pt4 including lesions in lung, liver, and soft tissue, not reported by the trial investigator. These lesions did not change in size or number over the course of treatment and were generally less than 1 cm in their largest dimension (no n- target lesions). These observations indicated that patients with predominant lymphatic disease but otherwise low visceral tumor burden can benefit from this immunotherapy. Moreover, they were consistent with the idea that further metastasis can be inhibited by this treatment.
EP10773491A 2009-10-23 2010-10-22 Krebsimmuntherapie und behandlungsverfahren Withdrawn EP2490713A2 (de)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US27962109P 2009-10-23 2009-10-23
US25465709P 2009-10-24 2009-10-24
US25585009P 2009-10-28 2009-10-28
PCT/US2010/053879 WO2011050344A2 (en) 2009-10-23 2010-10-22 Cancer immunotherapy and method of treatment

Publications (1)

Publication Number Publication Date
EP2490713A2 true EP2490713A2 (de) 2012-08-29

Family

ID=43415309

Family Applications (1)

Application Number Title Priority Date Filing Date
EP10773491A Withdrawn EP2490713A2 (de) 2009-10-23 2010-10-22 Krebsimmuntherapie und behandlungsverfahren

Country Status (7)

Country Link
US (1) US20110274723A1 (de)
EP (1) EP2490713A2 (de)
JP (1) JP2013508415A (de)
AU (2) AU2010310468A1 (de)
CA (1) CA2778707A1 (de)
MX (1) MX2012004721A (de)
WO (1) WO2011050344A2 (de)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013044169A1 (en) * 2011-09-21 2013-03-28 Nestec S.A. Methods for determining combination therapy with il-2 for the treatment of cancer
WO2013172926A1 (en) * 2012-05-14 2013-11-21 Yale University Immune biomarkers and assays predictive of clinical response to immunotherapy for cancer
EP2936157A1 (de) * 2012-12-21 2015-10-28 Servicio Andaluz De Salud Expression von beta2-mikroglobulin als prognostischer marker zur tumorimmunevasion und resistenz gegen krebsimmuntherapie und diagnostischer biomarker für patientenauswahl für spezifische gentherapie
AU2016229294B2 (en) 2015-03-06 2021-11-04 Beyondspring Pharmaceuticals, Inc. Method of treating cancer associated with a RAS mutation
US10155748B2 (en) 2015-07-13 2018-12-18 Beyondspring Pharmaceuticals, Inc. Plinabulin compositions
RU2753543C1 (ru) 2016-02-08 2021-08-17 Бейондспринг Фармасьютикалс, Инк. Композиции, содержащие тукаресол или его аналоги
WO2017194170A1 (en) * 2016-05-13 2017-11-16 Biontech Rna Pharmaceuticals Gmbh Methods for predicting the usefulness of proteins or protein fragments for immunotherapy
JP7025416B2 (ja) 2016-06-06 2022-02-24 ビヨンドスプリング ファーマシューティカルズ,インコーポレイテッド 好中球減少症を低減させるための組成物および方法
US11633393B2 (en) 2017-01-06 2023-04-25 Beyondspring Pharmaceuticals, Inc. Tubulin binding compounds and therapeutic use thereof
WO2018144764A1 (en) 2017-02-01 2018-08-09 Beyondspring Pharmaceuticals, Inc. Method of reducing neutropenia
SG11202006985TA (en) 2018-01-24 2020-08-28 Beyondspring Pharmaceuticals Inc Composition and method for reducing thrombocytopenia via the administration of plinabulin
MX2021012004A (es) * 2019-03-30 2021-11-04 Biontech Us Inc Composiciones y metodos para preparar composiciones de celulas t y usos de las mismas.
TW202241925A (zh) 2021-01-15 2022-11-01 德商英麥提克生物技術股份有限公司 用於不同類型癌症免疫治療的hla展示肽
US20230190807A1 (en) * 2021-10-06 2023-06-22 Immatics Biotechnologies Gmbh Tcr compounds, compositions, and methods of treating

Family Cites Families (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2606601A (en) 1948-12-14 1952-08-12 Knoll Associates Chair having a back rest in the form of a shell-like body
US5891955A (en) 1991-04-16 1999-04-06 Ibf Integrated Business And Finance S.A. Process for transforming a starting material containing at least two different thermoplastic materials into a new homogenous thermoplastic material
DE4143467C2 (de) 1991-05-17 1995-02-09 Max Planck Gesellschaft Peptidmotiv und dessen Verwendung
US6037135A (en) 1992-08-07 2000-03-14 Epimmune Inc. Methods for making HLA binding peptides and their uses
ES2276390T3 (es) 1992-11-05 2007-06-16 Sloan-Kettering Institute For Cancer Research Antigeno de membrana especifico de la prostata.
US5747271A (en) 1992-12-22 1998-05-05 Ludwig Institute For Cancer Research Method for identifying individuals suffering from a cellular abnormality some of whose abnormal cells present complexes of HLA-A2/tyrosinase derived peptides, and methods for treating said individuals
US5620886A (en) 1993-03-18 1997-04-15 Ludwig Institute For Cancer Research Isolated nucleic acid sequence coding for a tumor rejection antigen precursor processed to at least one tumor rejection antigen presented by HLA-A2
US5874560A (en) 1994-04-22 1999-02-23 The United States Of America As Represented By The Department Of Health And Human Services Melanoma antigens and their use in diagnostic and therapeutic methods
DE4423392A1 (de) 1994-07-04 1996-01-11 Birsner & Grob Biotech Gmbh Verfahren zur Identifizierung antigener Peptide
US5830753A (en) 1994-09-30 1998-11-03 Ludwig Institute For Cancer Research Isolated nucleic acid molecules coding for tumor rejection antigen precursor dage and uses thereof.
EP0812355A2 (de) 1995-02-28 1997-12-17 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Berlin Mittel zur therapie von tumoren und anderen hyperplasien
US5698396A (en) 1995-06-07 1997-12-16 Ludwig Institute For Cancer Research Method for identifying auto-immunoreactive substances from a subject
GB9711957D0 (en) 1997-06-09 1997-08-06 Isis Innovation Methods and reagents for vaccination
US6977074B2 (en) 1997-07-10 2005-12-20 Mannkind Corporation Method of inducing a CTL response
US6994851B1 (en) 1997-07-10 2006-02-07 Mannkind Corporation Method of inducing a CTL response
ES2265165T3 (es) 1997-07-10 2007-02-01 Mannkind Corporation Dispositivo para inducir una respuesta ltc.
US20030138808A1 (en) 1998-02-19 2003-07-24 Simard John J.L. Expression vectors encoding epitopes of target-associated antigens
US6709844B1 (en) 2000-11-16 2004-03-23 Mannkind Corporation Avoidance of undesirable replication intermediates in plasmid propagation
US8017590B1 (en) * 1999-10-22 2011-09-13 Sanofi Pasteur Limited Method of inducing and/or enhancing an immune response to tumor antigens
US20030215425A1 (en) 2001-12-07 2003-11-20 Simard John J. L. Epitope synchronization in antigen presenting cells
US6861234B1 (en) 2000-04-28 2005-03-01 Mannkind Corporation Method of epitope discovery
DE60219603T2 (de) 2001-02-26 2007-08-30 Kao Corp. Behälter
WO2002069907A2 (en) 2001-03-07 2002-09-12 Mannkind Corporation Anti-neovasculature preparations for cancer
WO2003008537A2 (en) 2001-04-06 2003-01-30 Mannkind Corporation Epitope sequences
DE10142351A1 (de) 2001-08-30 2003-03-20 Giesecke & Devrient Gmbh Initialisieren einer Chipkarte
ATE504594T1 (de) 2001-09-07 2011-04-15 Univ Johns Hopkins Med Sezernierte und zelloberflächengene, die in gutartigen und bösartigen kolorektaltumoren exprimiert werden
EP1453471B1 (de) 2001-11-07 2011-01-05 Mannkind Corporation Für epitope von antigenen kodierende expressionsvektoren und verfahren zu deren konzeption
US6706405B2 (en) 2002-02-11 2004-03-16 Analytical Services & Materials, Inc. Composite coating for imparting particel erosion resistance
US6715905B2 (en) 2002-06-17 2004-04-06 Birchwood Products Limited Lighting apparatus
EP1394066A1 (de) 2002-09-02 2004-03-03 Rockwool International A/S Transporteinheit mit einer Schutzverpackung und Verfahren zur Manipulierung dieser Verpackung
CN1691964A (zh) 2002-09-06 2005-11-02 曼康公司 表位序列
CA2529056C (en) 2003-06-17 2013-09-10 Mannkind Corporation Combinations of tumor-associated antigens in compositions for various types of cancers
ES2350043T3 (es) 2003-06-17 2011-01-17 Mannkind Corporation Composiciones para producir, incrementar y mantener las respuestas inmunes contra epítopos restringidos de mhc de clase i, con fines profilácticos o terapéuticos.
US20050028188A1 (en) 2003-08-01 2005-02-03 Latona Richard Edward System and method for determining advertising effectiveness
WO2005032677A1 (en) 2003-10-07 2005-04-14 Julian Jamison Kennedy Method of and apparatus for playing a card game
US20060159689A1 (en) 2004-06-17 2006-07-20 Chih-Sheng Chiang Combinations of tumor-associated antigens in diagnostics for various types of cancers
US20060008468A1 (en) 2004-06-17 2006-01-12 Chih-Sheng Chiang Combinations of tumor-associated antigens in diagnostics for various types of cancers
US20060057673A1 (en) 2004-06-17 2006-03-16 Liping Liu Epitope analogs
EP1773402A2 (de) 2004-06-17 2007-04-18 Mannkind Corporation Wirksamere immuntherapie mittels ergänzung diagnostischer verfahren mit therapeutischen verfahren
US7015608B2 (en) 2004-07-27 2006-03-21 Delco Remy International, Inc. Method and apparatus to suppress electrical noise in a rotor assembly for an electrical machine
EP1835932A2 (de) 2004-12-29 2007-09-26 Mannkind Corporation Verfahren zur auslösung, verbesserung und erhaltung von immunantworten gegen mhc-klasse-i-beschränkte epitope, für prophylaktische oder therapeutische zwecke
US20060153844A1 (en) 2004-12-29 2006-07-13 Thomas Kundig Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs
PL1833506T3 (pl) 2004-12-29 2016-01-29 Mannkind Corp Zastosowanie kompozycji zawierających różne antygeny związane z nowotworem jako szczepionek przeciwnowotworowych
JP2008526764A (ja) 2004-12-29 2008-07-24 マンカインド コーポレイション 免疫応答の誘導におけるcd4+細胞の回避方法
EP1890724A2 (de) * 2005-05-13 2008-02-27 Oxxon Therapeutics Limited Zusammensetzungen zur auslösung einer immunantwort
CN101273056A (zh) 2005-06-17 2008-09-24 曼康公司 表位类似物
WO2006138567A2 (en) 2005-06-17 2006-12-28 Mannkind Corporation Methods and compositions to elicit multivalent immune responses against dominant and subdominant epitopes, expressed on cancer cells and tumor stroma
EP3088400A1 (de) 2005-06-22 2016-11-02 Plexxikon Inc. Pyrrolo[2,3-b]pyridin-derivate als proteinkinasehemmer
JP4517984B2 (ja) 2005-09-01 2010-08-04 トヨタ自動車株式会社 ハイブリッド自動車
WO2008008541A2 (en) * 2006-07-14 2008-01-17 Mannkind Corporation Methods to elicit, enhance and sustain immune responses against mhc class-i restricted epitopes, for prophylactic or therapeutic purposes
US20080199458A1 (en) 2007-01-19 2008-08-21 Jian-Er Lin Influenza prevention and treatment composition
US20080199485A1 (en) 2007-02-15 2008-08-21 Mannkind Corporation Method for enhancing T cell response
US9469902B2 (en) 2014-02-18 2016-10-18 Lam Research Corporation Electroless deposition of continuous platinum layer
US9814289B2 (en) 2015-04-08 2017-11-14 Otter Products, Llc Protective folio case for an electronic device
EP3636238B1 (de) 2018-10-12 2021-04-28 Liko Research & Development AB Tore mit übergangsrampen für deckenhubschienen
CN113712788B (zh) 2021-08-13 2022-06-28 浙江益恒悦医疗科技有限公司 智能助行器的防摔控制方法、智能助行器、控制器

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2011050344A2 *

Also Published As

Publication number Publication date
US20110274723A1 (en) 2011-11-10
AU2016201722A1 (en) 2016-04-07
WO2011050344A3 (en) 2011-11-03
WO2011050344A2 (en) 2011-04-28
AU2010310468A1 (en) 2012-05-24
CA2778707A1 (en) 2011-04-28
MX2012004721A (es) 2012-06-25
JP2013508415A (ja) 2013-03-07

Similar Documents

Publication Publication Date Title
US20110274723A1 (en) Cancer immunotherapy and method of treatment
ES2351995T3 (es) Perfiles de antígenos asociados a tumores en el diagnóstico de cáncer e inmunoterapia.
AU2005321898B2 (en) Use of compositions comprising various tumor-associated antigens as anti-cancer vaccines
Guo et al. Therapeutic cancer vaccines: past, present, and future
Ophir et al. Personalized approaches to active immunotherapy in cancer
JP5283335B2 (ja) 各種癌の治療を目的とした腫瘍関連抗原の組合せ
CA2612512A1 (en) Combinations of tumor-associated antigens in diagnostics for various types of cancer
JP3857877B2 (ja) Ctl応答を誘導する方法
US20060159689A1 (en) Combinations of tumor-associated antigens in diagnostics for various types of cancers
Duperret et al. A designer cross-reactive DNA immunotherapeutic vaccine that targets multiple MAGE-A family members simultaneously for cancer therapy
JP2016532451A (ja) 前立腺癌処置のための組成物およびワクチン
MX2009000452A (es) Metodos para producir, mejorar y sostener respuestas inmunes contra epitopos restringidos mhc clase i para propositos profilacticos o terapeuticos.
Hirschowitz et al. Vaccines for lung cancer
Le et al. Next-generation cancer vaccine approaches: integrating lessons learned from current successes with promising biotechnologic advances
Izumoto Peptide vaccine
Fatima et al. Vaccines in Breast Cancer: Challenges and Breakthroughs. Diagnostics 2023, 13, 2175
Curti MART-1

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20120522

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20130408

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: MANNKIND CORPORATION

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20160105